You are on page 1of 27

REVIEW

published: 12 August 2021


doi: 10.3389/fphar.2021.699899

Traditional Uses, Phytochemical


Constituents and Pharmacological
Properties of Averrhoa carambola L.: A
Review
Fei Luan 1†, Lixia Peng 1†, Ziqin Lei 1, Xiyu Jia 1, Junbo Zou 2, Yan Yang 3, Xirui He 3* and
Nan Zeng 1*
1
State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional
Chinese Medicine, Chengdu, China, 2Department of Pharmacology, College of Pharmacy, Shaanxi University of Chinese
Medicine, Xianyang, China, 3Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China

Edited by:
Michał Tomczyk, Averrhoa carambola L. (star fruit) is an edible fruit that is extensively cultivated in
Medical University of Bialystok, Poland
southern China, Southeast Asia, India, and northern South America. It has a sweet
Reviewed by:
Ulrike Lindequist, and juicy taste and is frequently used in fruit salads and fruit platters, as a garnish in
University of Greifswald, Germany cocktail drinks and beverages, or squeezed into juice and served as a beverage.
Armando Caceres,
Traditionally, it has been used for treating diabetes and diabetic nephropathy,
Universidad de San Carlos de
Guatemala, Guatemala arthralgia, vomiting, lithangiuria, coughing, hangovers, and chronic paroxysmal
*Correspondence: headache for thousands of years. Currently, approximately 132 compounds have
Xirui He been isolated from A. carambola. Among them, flavonoids, benzoquinone, and their
xiruihe6105194@163.com
Nan Zeng glycosides have been considered as biologically active substances, which are
19932015@cdutcm.edu.cn responsible for various biological activities. Pharmacological studies have

These authors have contributed revealed that crude extracts or monomeric compounds from A. carambola
equally to this work
exhibit multiple bioactivities, such as anti-oxidant, anti-hyperglycemic, anti-
obesity, anti-hyperlipidemic, anti-tumor, anti-inflammatory, hepatoprotective,
Specialty section:
This article was submitted to cardioprotective, anti-hypertensive, neuroprotective, and others. Thus, A.
Ethnopharmacology, carambola is a valuable treatment in Chinese medicine with therapeutic potential
a section of the journal
Frontiers in Pharmacology for multiple diseases, especially diabetes and diabetes-related diseases. Even
Received: 24 April 2021 though it is a very promising candidate in the development of functional food
Accepted: 02 July 2021 and the pharmaceutical industry, reports on its bioactivities have only been
Published: 12 August 2021
conducted in vivo and in vitro and there is a gap in research regarding clinical
Citation:
settings and safety. This review therefore provides a comprehensive and systematic
Luan F, Peng L, Lei Z, Jia X, Zou J,
Yang Y, He X and Zeng N (2021) overview of current progress on botany, ethnopharmacology, phytochemistry,
Traditional Uses, Phytochemical pharmacology, and toxicity of A. carambola, providing a valuable reference for
Constituents and Pharmacological
Properties of Averrhoa carambola L.: further developments and applications of A. carambola in the pharmaceutical
A Review. industry and functional food.
Front. Pharmacol. 12:699899.
doi: 10.3389/fphar.2021.699899 Keywords: Averrhoa carambola, chemical composition, antidiabetic, anticancer, toxicology

Frontiers in Pharmacology | www.frontiersin.org 1 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

INTRODUCTION cultivated throughout tropical and warm subtropical areas


(Figure 2). The fruit is of high commercial value and is
Averrhoa carambola L., commonly known as carambola or star specially and extensively distributed and cultivated in southern
fruit, is a perennial tree in the family Oxalidaceae (Figure 1). It is China, Southeast Asia, India, and northern South America (Saikia
considered native to Malaysia, however, it is a tropical American et al., 2015; Leivas et al., 2016b; Varela-Martínez et al., 2019;
species introduced to Asia by the Spanish galleons and mainly Zulfajri et al., 2019). A. carambola is fleshy, crunchy, juicy,

FIGURE 1 | A. carambola: (A): whole plants; (B): fruits; (C): flowers and woods; (D): leaves (https://image.baidu.com/).

FIGURE 2 | Geographical distribution of A. carambola throughout the world (https://www.cabi.org/isc/datasheet/8082).

Frontiers in Pharmacology | www.frontiersin.org 2 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

slightly tart, acidic, and sweet in the taste. It is star-shaped and medicinal plants to ornamental plants in gardens. For instance, A.
golden-yellow in appearance and is frequently used in the carambola is widely planted as a decorative tree in the streets of
preparation of fruit salads and fruit platters, as a garnish in southern Chinese cities because of its beautiful appearance (Wu
cocktails and beverages, or squeezed into a juice and served as a et al., 2020b). More importantly, it is reported that the total
functional beverage. It is also used in jellies, ice creams, preserves, consumption per year of A. carambola in China is about 2.6
and sweets owing to its high moisture content and highly million tons, whereas the annual production of A. carambola is
perishability, especially in tropical regions such as Malaysia, only two million tons (Wu et al., 2020b).
Singapore, and Indonesia (Valim et al., 2016; Chua et al., To date, there has been no authoritative published systematic
2017; Huynh and Nguyen, 2017; Jia et al., 2018; Lu et al., and comprehensive review that focuses on all of the important
2018). For instance, in Malaysia star fruits are usually blended aspects of A. carambola. In the present review, recent advances in
with apples and braised with cloves and sugars or cooked along traditional uses, botanical characteristics, distribution, taxonomy,
with meat or seafood (Bhat et al., 2011). Generally, star fruits are phytochemical constituents, biological effects as well as the
regarded as an abundant source of various nutrients such as toxicities of A. carambola are comprehensively presented and
minerals, proteins, and vitamins, and also rich in natural critically evaluated. Furthermore, the underlying mechanism
phytochemicals such as flavonoids, terpenes, saponins, associated with the bioactivities of crude extracts or
alkaloids, proanthocyanidins, vitamin C, tartaric acid, oxalic components from this plant is also well summarized. The
acid, α-ketoglutaric acid, citric acid, vitamin B1 and B2, review is helpful for researchers by providing a comprehensive
carotene, pectin, cellulose, gallic acid, epicatechin, fatty acids, understanding of this increasingly important herb and provides a
volatile flavors, fibers, hemicellulose, polysaccharides, and sterols scientific basis for further study and exploitation of medicinal
(Shui and Leong, 2006; Benkeblia and Lopez, 2015; Leivas et al., agents or functional food from A. carambola in the future.
2015; Muthu et al., 2016; Yang et al., 2017; Zulfajri et al., 2019).
Simultaneously, GC-MS analysis has demonstrated that the
abundant fatty acids existing in A. carambola leaves were MATERIAL AND METHODS
α-linolenic acid (62.04%) and oleic acid (55.44%) in fruits.
Moreover, the proportion of total unsaturated fatty acids This review collected, analyzed, summarized literature on the
existing both in the fruits and leaves of A. carambola comprise botanical description, traditional uses, chemical constituents,
more than 77% of total fatty acid (Wei et al., 2014). The fructose pharmacological activities, and toxicities of A. carambola. All
content (38–48%) and glucose content (21–25%) have information was systematically gathered from globally accepted
predominantly sugar-based compositions in A. carambola ripe scientific databases by Internet databases, including Elsevier,
fruits, while sorbitol is also another major sugar alcohol ScienceDirect, PubMed, Web of Science, Wiley, Springer,
(2.4–10.5%) in ripe fruits (Ramadan et al., 2020). Additionally, SciFinder, ACS Publications, CNKI, WanFang, Google Scholar,
the presence of high amounts of fibers in this plant contributes to Baidu Scholar, The Plant List Database, and other literature
the absorption of glucose, restraint glucose diffusion into the sources (Ph.D. and MSc dissertations). All published
bloodstream, and maintain normal blood glucose levels (Fan contributions on A. carambola in different languages were
et al., 2020). Furthermore, the byproduct or pomace residue included and cited. The identification and examination of the
from A. carambola left after juice drink extraction contains collected works were based on titles and abstracts. The reference
more antioxidants than the extracted juice (Shui and Leong, lists of the retrieved publications were also checked to identify
2006). Interestingly, incorporation of 4% A. carambola fruit further relevant papers. The chemical structures of all isolated
juice and 6% Bambusa polymorpha Munro (Poaceae family) compounds were drawn by using ChemBioDraw Ultra 14.0
shoot extract, significantly prolonged the shelf life of pork software.
nuggets by at least 2 weeks (Thomas et al., 2016). Recent
studies found that an antifreeze protein purified from the cold
acclimated leaves of Drimys angustifolia Miers (Winteraceae BOTANICAL DESCRIPTION, GEOGRAPHIC
family) and synergistic pectin-maltodextrin-sodium chloride DISTRIBUTION, AND TAXONOMY
edible coating could dramatically increase the quality of frozen
A. carambola (Provesi et al., 2019; Mohd Suhaimi et al., 2021). Botanical Description
In recent years, phytochemical investigations have revealed Botanically, A. carambola is a medium-sized tree reaching up to
that the major chemical components of A. carambola mainly 3–15 m tall. The stem is gray bark. The leaf is odd-numbered
include flavonoids, terpenes, and other phenolics. Among them, compound leaves, alternate, leaflets 5-13, entire, ovoid, or elliptic,
2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione (122, 3–7 cm long, acuminate at apex, with a round base, skewed on
DMDD) is the most representative chemical compound with one side, sparsely pilose or glabrous underneath. The flower is
multiple biological activities (Gao et al., 2015; Xie et al., 2016; cymes or panicles; sepals 5, about 5 mm long, arranged in
Chen et al., 2017b). Pharmacological studies have demonstrated imbricate shape, synthetic ring at the base; petals slightly
that the crude extracts or active substances of A. carambola have dorsal, 0.8–1 cm long, purple-red, sometimes pink or white on
multiple health-promoting effects, and many of the biological the back; stamens 5-10; Ovary 5 compartments, many ovules in
effects above mentioned have ethnomedicinal uses. Furthermore, each compartment, style 5. The fruit is fleshy, drooping, with 5
the usable range of A. carambola is increasingly expanding from edges, rarely 6 or 3 edges, star-shaped in cross section, 5–8 cm

Frontiers in Pharmacology | www.frontiersin.org 3 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

long, light green or waxy yellow, sometimes dark red. The seed is a food supplement that can improve the appetite of people
dark brown (Flora of China Editorial Committee, 1998). with poor appetite (Vasconcelos et al., 2006; Soncini et al.,
2011). The traditional Indian medicine records that the ripe
Geographic Distribution fruits of A. carambola can be used for effectively curing the
A. carambola is traditionally considered to originate from hemorrhage of hemorrhoids and it is also regarded as a
Malaysia, although it has also been speculated to be a remedy for the treatment of eczema, fever, and diarrhea.
tropical American species introduced to Asia by the Spanish Furthermore, the ripe fruit of A. carambola is mainly
galleons. A. carambola has a wider climate range and can grow considered as digestive and tonic in Ayurveda (Vasant and
within the latitudinal range from 32°N to 30°S and withstand Narasimhacharya, 2014). As a Traditional Chinese Medicine
growing in both the hot humid tropics and subtropical (TCM), the roots, fruits, and leaves of A. carambola have been
countries including Egypt and Israel, and can tolerate short increasingly recognized as an effective herbal medicine in
periods of freezing temperatures as low as −3°C. It prefers well- invigorating kidney function and reinforcing Yang (it refers to
drained soils ideally between pH 5.5–6.5 but can tolerate pH the masculine, active and positive principle) and is used for
between 5 and 8.5 (Bircher and Bircher, 2000). In recent years, the treatment of various ailments with a long history (World
it has been extensively distributed and widely cultivated in Health Organization and Regional Office for the Western
most parts of the world (Figure 2), e.g., Asia countries Pacific, 2007; Wei et al., 2018). More specifically, the roots
including China and India, Africa countries including of A. carambola have been commonly accepted as a diuretic
Madagascar and Tanzania, North America countries and appetite stimulant agent, it is also used as an antidiarrheal
including Mexico and Honduras, Oceania countries and febrifugal drug with a long history of medical use in TCM
including Australia and French Polynesia, South America for the treatment of arthralgia, diabetes, DN, lithangiuria, and
countries including Brazil and Bolivia, etc. (https://www. chronic paroxysmal headache in ancient times (Cabrini et al.,
cabi.org/isc/datasheet/8082). 2011; Wen et al., 2013; Zheng et al., 2013; Chen et al., 2017b).
At the same time, A. carambola leaves have been commonly
Taxonomy utilized for alleviating vomiting, headaches, diabetes,
A. carambola belongs to the family Oxalidaceae, which coughing, and hangovers for a many years (Carolino et al.,
consists of over 900 species belonging to seven genera, such 2005; Ferreira et al., 2008). Furthermore, A. carambola fruits
as Dapania, Oxalis, Sarcotheca, Eichleria, Biophytum, are frequently applied to effectively remedy malarial
Hypseocharis, and Averrhoa. Among them, the genus splenomegaly and food poisoning caused by meat sources
Averrhoa mainly includes three species, namely A. (Pang et al., 2017). Overall, the leaves, roots, flowers, and
carambola, A. bilimbi L., and A. dolichocarpa Ruhayah and fruits of this plant, might be used as a dietary supplement and
Sunart (https://www.cabi.org/isc/datasheet/8082; Moresco should be further studied and developed as a functional food
et al., 2012). Importantly, A. carambola is commonly or therapeutic agent in the management of human health.
known as star fruit or carambola, bearing deeply ridged,
yellow-brown, edible fruit.
NUTRITIONAL AND PHYTOCHEMICAL
COMPOSITION
TRADITIONAL USES
Nutritional Composition
A. carambola has been traditionally used for thousands of Nutrient substances, such as minerals, vitamins, cellulose,
years in treating diabetes and diabetic nephropathy (DN), hemicelluloses, pectin, and others are contained in the fruit
arthralgia, vomiting, lithangiuria, coughing and hangovers, of A. carambola. It has been reported that A. carambola
and chronic paroxysmal headache. The different medicinal contains cellulose (60%), hemicelluloses (27%), and pectin
organs of A. carambola including leaves, roots, flowers, and (13%), which may contribute to controlling blood sugar
the fruits have been utilized as ethnomedicine in Chinese, levels (Lakmal et al., 2021). Moreover, carotene, vitamins,
Indian, Malaysian, and Brazilian medicine for a long time. For and acids were found in the ripe fruit of A. carambola, with
instance, the crushed shoots or leaves of A. carambola are high levels of vitamin C (25.8 mg/100 g fruit), tartaric acid
commonly applied in traditional Malaysia medicine to treat (4.37 mg/100 g fruit), vitamin B1 and B2 (0.12 mg/100 g fruit)
headache, chicken-pox, and ringworm, while a decoction of (Muthu et al., 2016). Furthermore, mineral elements were also
the leaves and fruits of A. carambola is generally used for found to be contained in A. carambola, with high levels of
treating vomiting, fevers, aphthous stomatitis, and angina potassium (167.13–168.0 mg/100 g fruit), phosphorous
(Yang et al., 2020b). In Sri Lanka, A. carambola fruits are (17.87–17.88 mg/100 g fruit), magnesium (11.85–12.05 mg/
traditionally used to treat and prevent diabetes mellitus due to 100 g fruit), calcium (6.37–6.40 mg/100 g fruit), sodium
their excellent hypoglycemic effects (Abeysekera et al., 2015). (3.8–3.85 mg/100 g fruit), iron (0.34–0.45 mg/100 g fruit),
In traditional Brazilian medicine, the fruit, juice, as well as tea zinc (0.29–0.51 mg/100 g fruit), copper (0.19–0.45 mg/100 g
made from leaves of A. carambola have been traditionally fruit), and manganese (0.04–0.52 mg/100 g fruit) (Muthu et al.,
utilized to prevent and treat diabetes, high blood pressure, and 2016). These results indicate that A. carambola is low-calorie
urinary system diseases and A. carambola is also considered as and may also have health-promoting properties.

Frontiers in Pharmacology | www.frontiersin.org 4 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 1 | Chemical components isolated and structurally identified from A. carambola.

No. Chemical constituents Molecular Extracts Parts References


formula

Flavonoids
1 Carambolaside R1 C35H38O14 EtOH Leaves Yang et al. (2020a)
2 Carambolaside R2 C35H38O14 EtOH Leaves Yang et al. (2020a)
3 Carambolaside R3 C35H38O15 EtOH Leaves Yang et al. (2020a)
4 Carambolaside S1 C41H48O18 EtOH Leaves Yang et al. (2020a)
5 Carambolaside S2 C41H48O18 EtOH Leaves Yang et al. (2020a)
6 Carambolaside T1 C41H48O18 EtOH Leaves Yang et al. (2020a)
7 Carambolaside T2 C41H48O18 EtOH Leaves Yang et al. (2020a)
8 Carambolaside T3 C41H48O19 EtOH Leaves Yang et al. (2020a)
9 3-Hydroxycarambolaside T1 C41H48O19 EtOH Leaves Yang et al. (2020a)
10 3-Hydroxycarambolaside P C47H58O24 EtOH Leaves Yang et al. (2020a)
11 Carambolaside M C32H42O18 EtOH Fruits Jia et al. (2018)
12 Carambolaside N C38H52O22 EtOH Fruits Jia et al. (2018)
13 Carambolaside O C47H58O23 EtOH Fruits Jia et al. (2018)
14 Carambolaside P C47H58O23 EtOH Fruits Jia et al. (2018)
15 Carambolaside Q C41H48O19 EtOH Fruits Jia et al. (2018)
16 Carambolaside A C21H24O9 MeOH Fruits Yang et al. (2015)
17 Carambolaside B C30H30O10 MeOH Fruits Yang et al. (2015)
18 Carambolaside C C27H34O14 MeOH Fruits Yang et al. (2015)
19 Carambolaside D C36H40O15 MeOH Fruits Yang et al. (2015)
20 Carambolaside E C27H34O13 MeOH Fruits Yang et al. (2016)
21 Carambolaside F C36H40O14 MeOH Fruits Yang et al. (2016)
22 Carambolaside G C36H40O14 MeOH Fruits Yang et al. (2016)
23 Carambolaside H C36H40O15 MeOH Fruits Yang et al. (2016)
24 Carambolaside I C41H48O18 MeOH Fruits Yang et al. (2016)
25 Carambolaside Ia C41H48O18 MeOH Fruits Yang et al. (2016)
26 Carambolaside J C47H58O22 MeOH Fruits Yang et al. (2016)
27 Carambolaside Ja C47H58O22 MeOH Fruits Yang et al. (2016)
28 8-carboxymethyl-(+)-epicatechin methyl ester C18H18O8 EtOH Fruits Jia et al. (2018)
29 (+)-Epicatechin C15H14O6 EtOH Fruits Jia et al. (2018)
30 Epicatechin-(5,6-bc)-4β-(p-hydroxyphenyl)-dihydro-2(3H)-pyranone C24H20O8 EtOH Leaves Yang et al. (2020b)
31 Epicatechin-(7,8-bc)-4α-(p-hydroxyphenyl)-dihydro-2(3H)-pyranone C24H20O8 EtOH Leaves Yang et al. (2020b)
32 Epicatechin-(7,8-bc)-4β-(p-hydroxyphenyl)-dihydro-2(3H)-pyranone C24H20O8 EtOH Leaves Yang et al. (2020b)
33 6-(S-2-Pyrrolidinone-5-yl)-epicatechin C19H19NO7 EtOH Leaves Yang et al. (2020b)
34 6-(R-2-pyrrolidinone-5-yl)-epicatechin C19H19NO7 EtOH Leaves Yang et al. (2020b)
35 (–)-Epicatechin C15H14O6 EtOAc Fruits Gunawardena et al. (2015)
36 Pinobanksin 3-O-β-D-glucoside C21H22O10 EtOH Fruits Jia et al. (2018)
37 Aromadendrin 3-O-β-D-glucoside C21H22O11 EtOH Fruits Jia et al. (2018)
38 Helicioside A C21H22O12 EtOH Fruits Jia et al. (2018)
39 Taxifolin 3′-O-β-D-glucoside C21H22O12 EtOH Fruits Jia et al. (2018)
40 Norathyriol C13H8O6 EtOH Fruits Jia et al. (2018)
41 Isorhamnetin 3-O-rutinoside C28H32O16 EtOH Fruits Jia et al. (2018)
42 Hovertichoside C C36H46O14 MeOH Fruits Yang et al. (2015)
43 Isovitexin 2″-O-α-L-rhamnopyranoside C27H30O14 MeOH Fruits Yang et al. (2015)
44 Carambolaflavone C27H30O13 MeOH Fruits Yang et al. (2015)
45 (+)-Catechin C15H14O6 Aqueous Roots Liao et al. (2019a)
46 Isovitexin C21H22O10 EtOH Leaves Araho et al. (2005)
47 Carambolaflavone A C27H30O13 EtOH Leaves Araho et al. (2005), Moresco et al. (2012),
Wang et al. (2018)
48 Carambolaflavone B C27H30O13 EtOH Leaves Araho et al. (2005), Moresco et al. (2012),
Wang et al. (2018)
49 Apigenin 6-C-(2′′-O-α-L-rhamnopyranosyl)-β-D-glucopyranoside C36H37O18 EtOH Leaves Moresco et al. (2012)
50 Cyanidin-3-O-β-D-glucoside C21H21O11Cl EtOH Leaves Gunasegaran (1992)
51 Cyanidin-3, 5-O-β-D-diglucoside C27H37O16Cl EtOH Leaves Gunasegaran (1992)
Terpenes
52 (5R,6S,7E,9R)-5,6,9-trihydroxy-7-megastigmene 9-O-β-D-glucoside C19H34O8 EtOH Fruits Jia et al. (2019)
53 Drovomifoliol C13H19O3 EtOH Fruits Jia et al. (2019)
54 3-oxo-α-ionol-9-O-β-D-glucoside C19H31O7 EtOH Fruits Jia et al. (2019)
55 Roseoside C19H30O8 EtOH Fruits Jia et al. (2019)
56 3-oxo-9-O-β-D-glucosyloxy-4,6E-megastigmadien C19H30O8 EtOH Fruits Jia et al. (2019)
57 4-oxo-β-ionol 9-O-β-D-glucoside C19H31O8 EtOH Fruits Jia et al. (2019)
58 Cannabiside D C19H30O9 EtOH Fruits Jia et al. (2019)
59 Dendranthemoside B C19H32O8 EtOH Fruits Jia et al. (2019)
(Continued on following page)

Frontiers in Pharmacology | www.frontiersin.org 5 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 1 | (Continued) Chemical components isolated and structurally identified from A. carambola.

No. Chemical constituents Molecular Extracts Parts References


formula

60 Icariside C27H30O11 EtOH Fruits Jia et al. (2019)


61 Officinoside A C19H32O8 EtOH Fruits Jia et al. (2019)
62 6S,7E,10S)-△9,15-10-hydroxyabscisic alcohol C15H22O4 EtOH Fruits Jia et al. (2019)
63 Abscisic acid C15H20O4 EtOH Fruits Jia et al. (2019)
64 Abscisyl β-D-glucoside C21H31O10 EtOH Fruits Jia et al. (2019)
65 9E-abscisic acid C15H20O4 EtOH Fruits Jia et al. (2019)
66 9E-abscisyl β-D-glucoside C21H32O10 EtOH Fruits Jia et al. (2019)
67 9E-abscisic alcohol β-D-glucoside C21H33O9 EtOH Fruits Jia et al. (2019)
68 cis-abscisic acid C15H20O4 EtOAc Fruits Gunawardena et al. (2015)
69 trans-abscisic acid C15H20O4 EtOAc Fruits Gunawardena et al. (2015)
70 trans-abscisic alcohol C15H22O3 EtOAc Fruits Gunawardena et al. (2015)
71 (6S,9R)-vomifoliol C13H20O3 EtOAc Fruits Gunawardena et al. (2015)
72 cis-abscisic acid β-D-glucopyranosyl ester C22H32O8 EtOAc Fruits Gunawardena et al. (2015)
73 trans-abscisic alcohol β-D-glucopyranoside C22H33O7 EtOAc Fruits Gunawardena et al. (2015)
74 (6S,9R)-roseoside C14H22O3 EtOAc Fruits Gunawardena et al. (2015)
75 cis-abscisic alcohol β-D-glucopyranoside C22H33O7 EtOAc Fruits Gunawardena et al. (2015)
76 Artemisinic acid C15H22O2 EtOH Fruits Yang et al. (2012)
77 3-β-hydroxyartemisinic acid C15H22O3 EtOH Fruits Yang et al. (2012)
78 Artemisinic acid 3-β-O-β-D-glucopyranoside C21H32O8 EtOH Fruits Yang et al. (2012)
79 3-β-hydroxyartemisinic acid β-D-glucopyranosyl ester C21H32O8 EtOH Fruits Yang et al. (2012)
80 Arjunolic acid C30H48O5 MeOH Fruits Yang et al. (2014)
Phenolics
81 Vanillic acid C 8H 8O4 MeOH Fruits Yang et al. (2014)
82 8,9,10-trihydroxythymol C10H10O4 MeOH Fruits Yang et al. (2014)
83 Carambolaside K C30H48O15 EtOH Fruits Jia et al. (2017)
84 Carambolaside L C32H52O16 EtOH Fruits Jia et al. (2017)
85 Koaburaside C14H20O9 EtOH Fruits Jia et al. (2017)
86 3,4,5-trimethoxyphenol-1-O-β-D-glucopyranoside C15H22O9 BuOH Roots Wen et al. (2012)
87 3,5-dimethoxy-4-hydroxyphenyl 1-O-β-apiofuranosyl (1’’→6’)-O-β-D- C19H28O13 BuOH Roots Wen et al. (2012)
glucopyranoside
88 3,4,5-trimethoxyphenyl 1-O-β-apiofuranosyl (1’’→6’)-β-glucopyranoside C20H30O13 BuOH Roots Wen et al. (2012)
89 Methoxyhydroquinone-4-β-D-glucopyranoside C13H18O18 BuOH Roots Wen et al. (2012)
90 3-hydroxy-4-methoxyphenol 1-O-β-D-apiofuranosyl-(1’’→6’)-O-β-D- C18H26O12 BuOH Roots Wen et al. (2012)
glucopyranoside
91 4-hydroxy-3-methoxyphenol 1-O-β-D-apiofuranosyl-(1’’→6’)-O-β-D- C18H26O12 BuOH Roots Wen et al. (2012)
glucopyranoside
92 Protocatechuic acid C 7H 6O4 EtOH Fruits Jia et al. (2017)
93 1-O-vanilloyl-β-D-glucose C14H18O9 EtOH Fruits Jia et al. (2017)
94 2,5-dimethoxy-3-undecylphenol C19H33O3 EtOAc Wood Chakthong et al. (2010)
95 5-methoxy-3-undecylphenol C18H31O2 EtOAc Wood Chakthong et al. (2010)
96 Gallic acid C 7H 6O5 Acetone Fruits Shui and Leong (2004)
Phenylpropanoids
97 Ferulic acid C10H10O4 MeOH Fruits Yang et al. (2014)
98 (+)-isolariciresinol 9-O-β-D-glucoside C27H36O12 EtOH Fruits Jia et al. (2017)
99 (+)-lyoniresinol 9-O-β-D-glucoside C28H38O13 EtOH Fruits Jia et al. (2017)
100 (−)-lyoniresinol 9-O-β-D-glucoside C28H38O13 EtOH Fruits Jia et al. (2017)
101 1-O-feruloyl-β-D-glucose C16H20O9 EtOH Fruits Jia et al. (2017)
102 Tarennanosides A C37H46O17 Aqueous Roots Liao et al. (2019a)
103 Fernandoside C36H44O16 Aqueous Roots Liao et al. (2019a)
104 7α-[(β-glucopyranosyl) oxy]-lyoniresinol C28H38O13 Aqueous Roots Liao et al. (2019a)
105 (+)-lyoniresinol 3α-O-β-D-glucopyranoside C28H38O13 Aqueous Roots Liao et al. (2019a)
106 (−)-lyoniresinol 3α-O-β-D-glucopyranoside C28H38O13 Aqueous Roots Liao et al. (2019a)
107 (−)-5′-methoxy-isolariciresinol 3α-O-β-D-glucopyranoside C27H36O12 Aqueous Roots Liao et al. (2019a)
108 (+)-5′-methoxy-isolariciresinol 3α-O-β-D-glucopyranoside C27H36O12 BuOH Roots Wen et al. (2012)
109 (+)-isolariciresinol 3α-O-β-D-glucopyranoside C26H34O11 BuOH Roots Wen et al. (2012)
110 (−)-isolariciresinol 3α-O-β-D-glucopyranoside C26H34O11 BuOH Roots Wen et al. (2012)
111 Reticulol C11H10O5 EtOAc Fruits Sritharan et al. (2019)
112 6-O-methyl-reticulol C11H10O5 EtOAc Fruits Sritharan et al. (2019)
113 5-methylmellein C11H12O3 EtOAc Fruits Sritharan et al. (2019)
114 7-hydroxy-5-methylmellein C11H12O4 EtOAc Fruits Sritharan et al. (2019)
Other constituents
115 Benzyl-1-O-β-D-glucopyranoside C13H18O6 BuOH Roots Wen et al. (2012)
116 (2S)-2-O-β-D-glucopyranosyl-2-hydroxyphenyl-acetic acid C14H18O8 BuOH Roots Wen et al. (2012)
(Continued on following page)

Frontiers in Pharmacology | www.frontiersin.org 6 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 1 | (Continued) Chemical components isolated and structurally identified from A. carambola.

No. Chemical constituents Molecular Extracts Parts References


formula

117 Methyl 2-β-D-apiofuranosyl-(1→6)-β-D-glucopyranosyloxybenzoate C19H26O12 EtOH Leaves Yang et al. (2020b)


118 Benzyl 2-β-D-apiofuranosyl-(1→6)-β-D-glucopyranosyloxybenzoate C25H30O12 EtOH Leaves Yang et al. (2020b)
119 Tecomin C15H20O9 EtOH Fruits Jia et al. (2017)
120 L-ascorbic acid C 6H 8O6 Acetone Fruits Shui and Leong (2004)
121 2-methoxy-6-nonyl-cyclohexa-2,5-diene-1,4-dione C16H24O3 EtOH Roots Wen et al. (2014)
122 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione C19H30O3 EtOH Roots Zhang et al. (2020)
123 5-O-methylembelin C18H28O4 EtOAc Woods Chakthong et al. (2010)
124 2-dehydroxy-5-O-methylembelin C18H28O3 EtOAc Woods Chakthong et al. (2010)
125 (+)-cryptosporin C14H12O6 EtOH Fruits Jia et al. (2017)
126 (1R*,3S*)-1-(5-hydroxymethylfuran-2-yl)-3-carboxy-6-hydroxy-8- C16H17NO6 MeOH Fruits Yang et al. (2014)
methoxyl-1,2,3,4-tetrahydroisoquinoline
127 (1S*,3S*)-1-methyl-3-carboxy-6-hydroxy-8-methyoxyl-1,2,3,4- C12H15NO4 MeOH Fruits Yang et al. (2014)
tetrahydroisoquinoline
128 Heptyl vicianoside C18H34O10 EtOH Fruits Yang et al. (2019b)
129 Octyl vicianoside C21H30O11 EtOH Fruits Yang et al. (2019b)
130 cis-3-hexenyl rutinoside C34H43O16 EtOH Fruits Yang et al. (2019b)
131 Methyl 2-O-β-D-fucopyranosyl-α-L-arabinofuranoside C12H22O9 EtOH Fruits Yang et al. (2019b)
132 Methyl α-D-fructofuranoside C7H14O6 EtOH Fruits Yang et al. (2019b)

EtOH, ethanol; EtOAc, ethyl acetate; n-BuOH, n-butanol.

Phytochemical Compounds Terpenes


Currently, approximately 132 phytochemical compounds have Terpenes are a group of secondary metabolites in plants that
been separated and identified from A. carambola, which contains consist of one or more isoprene subunits (Bahramsoltani et al.,
flavonoids, terpenes, phenylpropanoids, and their glycosides, 2020). They have the function of promoting the coloring in many
among others. These include flavonoids, benzoquinone and and various vegetables and fruits (Farias et al., 2020). To date, 29
their glycosides, which have been considered as the terpenes (52–80) have been mainly separated and identified from
biologically active components responsible for multiple the fresh fruits of A. carambola. Gunawardena et al. (2015)
bioactivities. The compounds isolated from A. carambola are analyzed different terpenes in star fruits using NMR and MS
documented and listed in Table 1 and the chemical structures are methods, and the major terpenes identified were cis-abscisic acid
drawn and presented in Figure 3. (68, 12 mg), trans-abscisic acid (69, 3.5 mg), trans-abscisic
alcohol (70, 12 mg), (6S,9R)-vomifoliol (71, 8.5 mg), cis-
Flavonoids abscisic acid β-D-glucopyranosyl ester (72, 19 mg), trans-
Various studies have found that the flavonoids isolated from this abscisic alcohol β-D-glucopyranoside (73, 12 mg), (6S,9R)-
plant possess excellent antioxidant and radical scavenging roseoside (74, 12 mg), and cis-abscisic alcohol β-D-
properties, which can be used to prevent and treat the glucopyranoside (75, 113 mg). Moreover, Jia et al. (2019)
occurrence of chronic and cardiovascular illness (Maqsood found that the terpenes-derived components from star fruits
et al., 2020). Until now, 51 flavonoids (1–51) have been are primarily C13- and C15-norisoprenoids, which
separated and characterized by nuclear magnetic resonance tremendously strengthen the flavor of A. carambola fruits.
(NMR) and mass spectrometer (MS) technologies from the Information of these terpenes is listed in Table 1. The
leaves, fruits, and roots of A. carambola. Among these are chemical structures were draw by ChemBioDraw Ultra 14.0
compounds (1–27) that are dihydrochalcone C-glycosides and and are shown in Figure 3. However, the pharmacological
other compounds (28–35) are flavan-3-ols, of which all exhibited activities of most terpenes is still unclear.
significant radical scavenging activities against the DPPH and
ABTS, while some compounds (36–51) are other types with Phenolics
multiple structures. Both the compounds carambolaflavone A Phenolic compositions are represented as one of the major classes
(47) and carambolaflavone B (48) showed excellent of plant secondary metabolites and extensively dispersed among
antihyperglycemic activity both in hyperglycemic and diabetic plant parts, phytochemical studies have found that these
rats’ model (Cazarolli et al., 2009; Cazarolli et al., 2012). compounds principally exist in the roots and fruits of A.
Afterward, Wang et al. (2018) conducted the total synthesis of carambola (Wen et al., 2012; Yang et al., 2014; Jia et al., 2017;
the enantiomers of carambolaflavone A (47) and found that Liao et al., 2019a). At present, 16 phenolic components (81–96)
structurally the β-fucosyl moiety absolute configuration was D were isolated and characterized by FT-IR, 1H-NMR, and
instead of L. Information of these isolated flavonoids is listed in 13
C-NMR, from the roots and fruits of A. carambola with
Table 1. Their chemical structures were drawn by ChemBioDraw excellent antioxidant properties. Among them, compounds 83,
Ultra 14.0 and are described in Figure 3. 84, 94, and 95 are alkyl phenols in structure. Pang et al. (2016)

Frontiers in Pharmacology | www.frontiersin.org 7 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

FIGURE 3 | Chemical structures of compounds isolated from A. carambola.

Frontiers in Pharmacology | www.frontiersin.org 8 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

FIGURE 3 | (Continued).

Frontiers in Pharmacology | www.frontiersin.org 9 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

FIGURE 3 | (Continued).

compared the phenolic and flavonoid compound content of four been found in A. carambola fruit, compounds reticulol (111) and
popular cultivars of A. carambola that originated from southern 6-O-methyl-reticulol (112) are isocoumarins in structure, 5-
China and found that the contents of bound, free, and total methylmellein (113) and 7-hydroxy-5- methylmellein (114)
phenolic for four cultivars were 6.4–19.7, 162.5–286.8, and are dihydroisocoumarins in structure. Reticulol (111)
174.5–293.1 mg gallic acid equivalents per 100 g fresh weight, displayed moderate antioxidant capacity against DPPH with
respectively. The contents of bound, free, and total flavonoid of the IC50 value of 58 μg/ml (Sritharan et al., 2019). These
the four cultivars were 1.1–7.8, 100.7–234.0, and 104.4–235.1 mg phenylpropanoid constituents are summarized in Table 1 and
catechin equivalents per 100 g fresh weight, respectively, which the corresponding chemical structures were also draw by
indicated that a certain amount of non-flavonoid phenolic ChemBioDraw Ultra 14.0 and are presented in Figure 3.
substances exist. These phenolic compositions are summarized
in Table. Their chemical structures were drawn by ChemBioDraw Other Compounds
Ultra 14.0 and are presented in Figure 3. Up to date, apart from the chemical compounds listed above, few
compounds (115–132) have been investigated and summarized
Phenylpropanoids in Table 1 and the corresponding chemical structures were
Phenylpropanoids are a kind of plant-derived organic compound, presented in Figure 3. Briefly, compounds 117 and 118 are
and these compounds are mainly derived from phenylalanine and identified as benzoic acid, compounds 121–125 are quinones,
tyrosine (Yao et al., 2020). At present, 18 phenylpropanoids compounds 126 and 127 are tetrahydroisoquinoline alkaloids,
(97–114) have been successfully separated and chemically and components 128–132 are identified as alkyl glycosides.
identified by analyses of spectroscopic data including 1H-NMR Among them, 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-
and 13C-NMR, from the fruits and roots of A. carambola. These dione (122) was obtained and characterized from A.
compounds can be classified as simple phenylpropanoids, carambola roots showing multiple bioactive properties both in
lignans, and coumarins based on their substructure type. cell and animal experimental studies, including anti-cancer (Gao
Among them, four simple phenylpropanoids (97–101) have et al., 2015; Chen et al., 2017b), anti-diabetic nephropathy (Lu
been reported for the A. carambola fruit, twelve lignans et al., 2019), anti-obesity (Li et al., 2016), anti-hyperglycemic
(102–110) were primarily achieved and identified from A. (Zhang et al., 2020), anti-inflammatory (Xie et al., 2016), and
carambola roots. Furthermore, four coumarins (111–114) have neuroprotective activities (Wei et al., 2018).

Frontiers in Pharmacology | www.frontiersin.org 10 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

PHARMACOLOGICAL ACTIVITIES features, and insights into the underlying mechanisms have
also been provided, although they are not yet fully understood.
Antioxidant Activity The underlying mechanism of the anti-hyperglycemic activity of
Among pharmacologically active substances, natural antioxidants crude extracts or bioactive substances from A. carambola is
have gained widespread attention because they are safe and have presented in Table 2 and Figure 4.
low toxicity, and promising biological functions (Rufino et al.,
2011). A. carambola leaves (ACL) supposedly exhibit the most Crude Extract
potent antioxidant activities determined by DPPH, FRAP, and Evaluations of the hypoglycemic activity of A. carambola crude
TEAC assays (Table 2). Phytochemical studies have shown that extracts on rats and mice have been conducted with encouraging
these leaves are rich in phenolic and flavonoid components, results. In one such assessment, mice with STZ-induced diabetes
which are closely associated with antioxidant effects; this given root extracts of A. carambola (REAC at daily doses of 150,
indicates that ACL is potentially an abundant source of 300, 600, and 1,200 mg/kg for 21 days) orally had significantly
natural antioxidants and could help prevent and treat decreased blood glucose, TC, TGs, and FFAs levels and elevated
oxidative stress-related diseases (Chen et al., 2017a). In a insulin content in their serum. Mechanically, REAC markedly
systematic comparison between twenty locally available fruits downregulated pro-apoptosis caspase-3/8/9 and Bax protein
planted in Sri Lanka, Silva and Sirasa (2018) demonstrated that A. expressions and upregulated anti-apoptotic Bcl-2 protein
carambola had the third most potent antioxidant properties based expression. Additionally, REAC prevented pancreatic β cell
on assays against FRAP and DPPH activities, total flavonoid apoptosis in these mice (Xu et al., 2014). The findings of that
content, total phenolic content, and vitamin C content. Siddika investigation suggest that REAC possesses remarkable
et al. (2020) found that the methanol extract of A. carambola hyperglycemic abilities that could improve metabolic functions
leaves (MEACL, at 50–375 μg/ml) demonstrated dose-dependent and suppress the apoptosis of pancreatic β cells.
moderate antioxidant activity when assayed against DPPH and Research into the anti-hyperglycemia activities of REAC on
ABTS+, with IC50 values of 62.0 and 6.0 μg/ml, respectively. Other regulating the TLR4/NF-κB signaling pathway in STZ-induced
phytochemical investigations have shown MEACL is rich in diabetic mice showed that REAC (at daily doses of 300, 600, and
phenolics, which could be the drive behind its radical 1,200 mg/kg, i.g. for 14 days) decreased the serum contents of
scavenging activity. Using paper spray ionization (PSI) fasting blood glucose (FBG), TNF-α, and IL-6 significantly and
coupled to high-resolution mass spectrometry, one study downregulated TLR4 and NF-κB protein and mRNA expressions
revealed that a bioactive compound, norathyriol, isolated from in pancreatic tissue (Xu et al., 2015). In their assessment of the
ethanol extracts of the bark of A. carambola (EEBAC) had protective features of REAC on renal function injury in STZ-
antioxidant properties. EEBAC (at concentrations of 1.0, 3.0, stimulated diabetic mice, Xu et al. (2017) found that REAC
10, 30, and 100 μg/ml) displayed concentration-dependent treatment reduced FBG, blood urea nitrogen (BUN), and
antioxidant characteristics via suppressing the activities of creatinine levels in serum significantly, strengthened SOD,
α-glucosidase, elastase, ABTS+, DPPH, and tyrosinase enzyme, GSH-Px, and CAT activities considerably, and lessened MDA
with IC50 values of 7.15, 20.34, 26.29, 55.55, and 56.46 μg/ml, levels and Cyto-C, AIF, and caspase-3 protein expressions in the
respectively (Islam et al., 2020). Compound (40) at 2.5, 5, and kidney tissues of the mice.
10 μg/ml, has shown powerful antioxidant effects against DPPH After orally treating fluoride-induced hyperglycemia,
and ABTS+, with corresponding IC50 values of 4.9 and 9.63 μg/ml hypercholesterolemia, and oxidative stress in rats with fruit
(Islam et al., 2020). The efficiency and potency of crude extracts powder from A. carambola (FPAC, at daily doses of 2.5, 5.0,
or bioactive ingredients from A. carambola suggest it is a and 10 g for 30 days), the plasma glucose level and G-6-pase,
promising antioxidant in the pharmaceutical and functional SGOT, SGPT, ACP, and ALP activities reduced notably, while the
food industries. activities of hepatic glycogen, hexokinase, and FPAC increased
dose-dependently. Increments in the atherogenic index of plasma
Anti-Hyperglycemic Activity (AIP), total lipids (TL), TC, TG, LDL-C, and VLDL-C in plasma,
Diabetes mellitus is characterized by a chronic hyperglycemic TL, TC, and TG in the liver and the decrease in HDL-C were also
condition possibly induced by insulin deficiency, damage to reversed. Additionally, FPAC prompted an increase in CAT,
insulin signaling, or non-autoimmune etiology or caused by a SOD, GSH-Px, GSH, and total ascorbic acid (TAA) activities
remarkably diminished insulin sensitivity (Teng et al., 2018). This significantly, and a decline in MDA content in the hepatic and
disorder poses a significant public health care problem, with its renal tissues of rats (Vasant and Narasimhacharya, 2014).
prevalence continuing to rise globally (Chen et al., 2019). Type 2 STZ-induced diabetic mice receiving juice extracts from A.
diabetes mellitus, in particular, is primarily distinguished by carambola (JEAC) intragastrically (at daily doses of 25, 50, and
insulin resistance and β cell dysfunction, which cause insulin 100 mg/kg for 21 days) have displayed markedly reduced FBG,
secretion reduction (Uuh-Narváez et al., 2021). Diabetes mellitus FFA, MDA, TC, TG, serum creatinine (Scr), and BUN levels and
patients can die if they develop diabetic kidney disease. significantly increased insulin, sorbitol dehydrogenase (SDH),
In the past 5 years, several studies have extensively explored cAMP, and SOD activities. In the same study, diabetes-instigated
the antidiabetic potential of A. carambola using various changes in kidney tissues, including thickened and tubular
experimental models. The findings have revealed that A. basement membranes and glomerular hypertrophy, greatly
carambola and its glycosides exhibit outstanding antidiabetic improved, and the expressions of related mRNAs and

Frontiers in Pharmacology | www.frontiersin.org 11 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 2 | Pharmacological effects of crude extracts and bioactive compounds of A. carambola.

Pharmacological Compounds/ Types Testing subjects Doses/Duration Effects/Mechanisms References


activity Extracts

Antioxidant activity
MEACL In vitro DPPH and ABTS+ assays 50–375 μg/ml Showed moderate free radical Siddika et al. (2020)
scavenging activity against
DPPH and ABTS+ free radical
with the IC50 were 62.0 and
6.0 μg/ml, respectively
PRAC In vitro DPPH and ABTS+ assays 0.5–2.5 mg/ml Showed significant free radical Liao et al. (2019b)
scavenging activity against
DPPH and ABTS+ with the IC50
of 0.10 and 0.33 mg/ml,
respectively
EEBAC In vitro α-glucosidase, elastase, ABTS+, 1.0, 3.0, 10, 30, and Showed significant antioxidant Islam et al. (2020)
DPPH, and tyrosinase enzyme 100 μg/ml activity, and the IC50 values
assays were 7.15, 20.34, 26.29, 55.55
and 56.46 μg/ml, respectively
40 In vitro DPPH and ABTS+ assays 2.5, 5, and 10 μg/ml Showed significant radical Islam et al. (2020)
scavenging activity against the
DPPH and ABTS+ with the IC50
values of 4.9 and 9.63 μg/ml,
respectively
Anti-hyperglycemic activity
REAC In vivo STZ-induced diabetic mice 150, 300, 600, and Blood glucose, TC, TGs, and Xu et al. (2014)
1,200 mg/kg, daily for FFAs levels ↓; insulin level ↑;
21 days caspase-3, caspase-8,
caspase-9, and Bax protein
expressions ↓; Bcl-2 protein
expression ↑
REAC In vivo STZ-induced diabetic mice 300, 600, and FBG, IL-6 and TNF-α levels ↓; Xu et al. (2015)
1,200 mg/kg, i.g., daily TLR4 and NF-κB mRNA and
for 14 days protein expression
REAC In vivo STZ-induced diabetic mice 300, 600, and FBG, Cr, BUN, and MDA levels Xu et al. (2017)
1,200 mg/kg, i.g., daily ↓; SOD, GSH-Px, CAT activities
for 42 days ↑; Cyto-C, AIF, and caspase-3
protein expressions ↓
FPAC In vivo Fluoride-induced hyperglycemia, 2.5, 5.0, and 10 g, i.g., Blood glucose, G-6-pase, Vasant and
hypercholesterolemia, and for 30 days SGOT, SGPT, ACP, and ALP Narasimhacharya
oxidative stress of rat’s model levels ↓; hepatic glycogen and (2014)
hexokinase, and FRAP
activities ↑; plasma AIP, TL, TC,
TG, LDL-C, VLDL-C, hepatic
lipids-TL, TC and TG levels ↓;
HDL-C level ↑; CAT, SOD, GPx,
GSH, TAA activities ↑; MDA
level ↓
JEAC In vivo STZ-induced diabetic mice 25, 50, and 100 mg/kg, FBG, FFA, TC, TG, Scr, BUN, Pham et al. (2017)
i.g., once a day for and MDA levels ↓; SDH, cAMP,
21 days SOD, and insulin activities ↑;
CTGF and TGF-β1 mRNA and
protein expressions ↓
JEAC In vivo STZ-induced diabetic mice 5, 10, and 20 g/kg, FBG, blood glucose, area Yang et al. (2019a)
orally, once a day for under curve, LDH, GC and
14 days pyruvate ↓; FINS level ↑
TFACL In vivo Alloxan-induced diabetic mice 0.2, 0.4, and 0.8 g/kg, FBG level ↓; glucose Liu et al. (2013)
and STZ-induced diabetic rats i.g., daily for 7 days tolerance ↑
EEACB In vivo STZ-induced diabetic mice 50 and 100 mg/kg, i.g., Blood glucose level ↓ Islam et al. (2020)
for three successive
days
47 In vivo STZ-induced diabetic rats 20 and 50 mg/kg, i.g., Serum blood glucose level ↓; Cazarolli et al. (2012)
48 for 3 h insulin secretion ↑
48 In vitro 14
C-glucose uptake in rat soleus 50 and 100 μM for 1 h Glucose uptake and glucose Cazarolli et al. (2009)
muscle transport ↑
(Continued on following page)

Frontiers in Pharmacology | www.frontiersin.org 12 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 2 | (Continued) Pharmacological effects of crude extracts and bioactive compounds of A. carambola.

Pharmacological Compounds/ Types Testing subjects Doses/Duration Effects/Mechanisms References


activity Extracts

105 In vivo STZ-induced diabetic mice 20, 40, and 80 mg/kg, FBG, FINS, and ISI levels ↓; NF- Wen et al. (2013)
106 i.g., daily for 14 days κB, caspase-3, caspase-8,
caspase-9, and Bax protein
expressions ↓
121 In vivo STZ-induced diabetic mice 30, 60, and 120 mg/kg, FBG, TC, TG, FFA, GHb, FINS, Qin et al. (2019)
i.g., once daily, for MCP-1, TNF-α, IL-6 and MDA
21 days levels ↓; SOD, GSH activities ↑
121 In vivo STZ-induced diabetic mice 30, 60, and 120 mg/kg, TLR4, MyD88, p-NF-κB, TNF- Qin et al. (2020)
i.g., once daily, for α, and IL-6 mRNA and protein
21 days expression levels ↓
122 In vivo Type 2 diabetic KKAy mice 12.5, 25, and 50 mg/kg, FBG, AGEs glycosylated Zheng et al. (2013)
i.g., daily, for 56 days protein and TC levels ↓; albumin
level ↑
122 In vivo Type 2 diabetic KKAy mice 12.5, 25, and 50 mg/kg, FBG level ↓; RAGE, NF-B, TGF- Zheng et al. (2021)
i.g., daily, for 56 days β1 and CML protein expression
levels ↓
122 In vivo DN model established by STZ in 12.5, 25, and 50 mg/kg, TC, TG, HDL, LDL, Scr, BUN, Lu et al. (2019)
TLR4 knockout mice and wild- i.g., daily for 28 days and blood glucose ↓; IL-6 and
type mice TNF-α level ↓; TLR4, MyD88
and NF-κB mRNA and protein
expressions ↓
122 In vitro HG-induced HK-2 cells 30 μM for 48 h Blood glucose ↓; Vimentin Zhang et al. (2019)
mRNA and protein level ↑;
TLR4 and E-cadherin mRNA
and protein levels ↓; BAMBI ↑;
Smad2/3 ↓
122 In vivo Diabetic kidney disease mice 12.5, 25, and 50 mg/kg, TC, TG, LDL-C, FBG, CysC, Zhang et al. (2020)
model induced by Wild type and i.g., once daily, for and urinary albumin levels ↓;
TLR4 knockout 28 days TLR4, TGF-β1 and Smad2/3
mRNA and protein levels ↓
Antihyperlipidemic activity
IFRF In vivo Murine model Diets formulations, i.g., TG, TC, HDL, and LDL levels ↓ Herman-Lara et al.
FF for 30 days (2014)
MEACL In vivo Poloxamer-407-induced NM TC, TG, LDL-C, VLDL-C and AI Saghir et al. (2016)
hyperlipidemic rat model levels ↓
MEACL In vivo HFD-induced hyperlipidemic rats 250, 500, and TC, TG, LDL-C, VLDL-C, and Aladaileh et al. (2019)
1,000 mg/kg, i.g., daily, AI ↓; HDL-C ↑; GSH, GPx,
for 35 days SOD, CAT activities ↑; MDA
level ↓
Anti-obesity activity
CEPAC In vitro 3T3-L1 preadipocytes 10, 100, 500, and TG accumulation ↓; PPAR-γ Rashid et al. (2016)
1,000 mg/ml and C/EBPα mRNA
expressions ↓; PPAR-α mRNA
expression ↑
122 In vivo high-fat diet (HFD) in mice 12.5, 25, and 50 mg/kg, BW and adipose tissue Li et al. (2016)
i.g., daily for 28 days weights, blood glucose, insulin,
TC, TG, FFA, IL-6, TNF-α levels
↓; TLR4 and MyD88
expressions ↓; insulin
secretion ↑
Antitumor activity
ACE In vivo DENA-induced and CCl4- 25 mg/kg, i.g., for five Tumor incidence, tumor yield, Singh et al. (2014)
promoted liver cancer in mice consecutive days tumor burden ↓; LPO level ↓;
GSH, SOD, CAT, total proteins
content activities ↑
MEACL In vivo EAC cell bearing mice 25 and 50 mg/kg, i.g., viable cells and body weight ↓; Siddika et al. (2020)
for 5 days survival time ↑; Hgb, WBC,
RBC numbers ↑; p53 and Bax
protein expression ↑
122 In vitro Human breast cancer MCF-7 and 10, 32, 100 μM for 24 h Caspase-3/7, -8, and -9 Gao et al. (2015)
BT20 cells activities ↑; TRAIL-R1, TRAIL-
R2, Bad, and BID protein
expressions ↑; cIAP, XIAP, and
(Continued on following page)

Frontiers in Pharmacology | www.frontiersin.org 13 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 2 | (Continued) Pharmacological effects of crude extracts and bioactive compounds of A. carambola.

Pharmacological Compounds/ Types Testing subjects Doses/Duration Effects/Mechanisms References


activity Extracts

Survivin protein expressions ↓;


G1 phase cell cycle arrest, ROS
↑; NF-κB ↓
122 In vivo Transplanted 4T1 breast cancer 25, 50 and 100 mg/kg, Survival time ↑; tumor growth ↓; Chen et al. (2017b)
cells bearing mouse i.g., for 14 days TNF-α, IL-6, IL-12, TGF-β,
VEGF ↓; Bax, cleaved
caspases-3 and -9 ↑; Bcl-2,
MMP-2 and -9, NF-κB and
IκBα ↓
122 In vitro Radio-sensitivity of 4T1 breast 100 μM for 2 or 24 h TIE, TRD ↓; radio-sensitivity of Muhammed et al.
carcinoma cell lines the 4T1 cells ↑ (2019)
122 In vitro Lung cancer H1299 cells 4.0, 6.0, and 8.0 μg/ml Cell apoptosis ↑; ERK/MAPK ↓; Zhou et al. (2019)
for 24 h inhibition rates were 22.50,
30.13, and 58.87%,
respectively
122 In vivo Hepatocarcinoma in nude mice 25, 50, and 100 mg/kg, Tumor weight ↓; liver and Wu et al. (2020a)
i.g., daily for 12 days spleen indexes ↓; IL-2 and IL-
10 levels ↓; inhibition rates were
66.39, 63.11, and 47.33%,
respectively; WBC, HGB, and
PLT numbers ↑; TLR4, MyD88,
and NF-κB expressions ↓
Anti-inflammatory activity
EEACL In vivo Croton oil-induced mouse ear 0.03–1.0 mg/ear Edema (IC50: 0.05) ↓; MPO Cabrini et al. (2011)
edema activity (IC50: 0.22) ↓
PFSCW In vivo Adult female Swiss mice 100 and 300 mg/kg Paw edema ↓ Leivas et al. (2016a)
122 In vivo Pancreatic β-cell line Min6 cells 10, 15, and 20 μmol/L TNF-α, IL-6 and MCP-1 level ↓; Xie et al. (2016)
cleaved-caspase-3, -8 and -9,
TLR4, MyD88, and NF-KB
expressions ↓; Bcl-2/Bax
ratio ↑
Hepatoprotective activity
FPEAC In vivo Leptin receptor- deficient (db/db) 10, 20, and 30 g/kg, i.g., TG, TC, LDL-C, and NEFA level Pang et al. (2017)
mice daily, for 8 weeks ↓; AST and ALT activities ↓;
HDL-C level ↑; p-AMPK protein
expression ↑; SREBP-1c, FAS
and SCD1 mRNA and protein
expression levels ↓; mircoRNA-
34a and mircoRNA-33
expression levels ↓
FJAC In vivo STZ-induced diabetic mice 5, 10, and 20 g/kg, i.g., MAD and cAMP levels ↓; SDH, Yang et al. (2018)
daily for 14 days MDA, and SOD activities ↑
EACR In vivo CCl4-induced acute liver injury in 0.3, 0.6, and 1.2 g/kg, f AST, ALT, IL-1, IL-6, MDA Huang et al. (2019)
mice i.g., dialy, for 7 days levels ↓; SOD, GSH, GSH-Px
activities ↑; TNF-α, NF-κB,
caspase-3 protein expression
levels ↑
EACR In vivo Liver fibrosis (HF) rats induced by 0.25, 0.5, and 1.0 g/kg, Albumin/globulin (A/G) ratio ↑; Liang et al. (2020b)
CCl4 i.g., dialy, for 28 days TBIL and TC levels ↓; NF-κB
and Bax expression levels ↓;
Bcl-2 expression level ↑
EACR In vivo CCl4-induced acute liver injury in 0.25, 0.5, and 1.0 g/kg, AST, ALT, AKP, Hyp, HA, LN, Huang et al. (2020)
rats i.g., dialy, for 8 weeks Col III, Col IV, MDA levels ↓;
SOD and GSH-Px activities ↑;
COL-1a1, α-SMA, TIMP2,
TGF-β1, Smad-2 and Smad-4
mRNA expression levels ↓;
α-SMA, TIMP2, TGF-β1,
Smad-2, Smad-3 and Smad-4,
Bax and cleaved caspase-3
proteins expression levels ↓;
Smad-7 mRNA expression and
(Continued on following page)

Frontiers in Pharmacology | www.frontiersin.org 14 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

TABLE 2 | (Continued) Pharmacological effects of crude extracts and bioactive compounds of A. carambola.

Pharmacological Compounds/ Types Testing subjects Doses/Duration Effects/Mechanisms References


activity Extracts

Smad-7 and Bcl-2 protein


expression ↑
Cardioprotective activity
AEAC In vivo Rats with ventricular remodelling 50, 100, and TGF-β, Ang II, iNOS, ECE, ET-1 Liang et al. (2020a)
induced by isoprenaline 200 mg/kg, i.g., daily, for levels and expressions ↓; VR
14 days index, CVF ↓; tNOS, eNOS
protein expression levels ↑
Antihypertensive activity
AELAC In vivo Normotensive rats 12.5–50.0 mg/kg, i.v. MAP ↓; Ca2+-free medium ↓ Soncini et al. (2011)
EEACR In vivo Normal rats 150, 300, and Blood pressure ↓ Tang et al. (2017)
600 mg/kg, i.g.
FACF In vivo Normal rats and rats with 300, 600, and SBP, DBP, MBP, blood Huang et al. (2017)
hypertension induced by L-NAME 1,200 mg/kg, i.g., daily, pressure ↓
for 5 weeks
Neuroprotective activity
122 In vivo APP/PS1 transgenic AD mice 12.5, 25, and 50 mg/kg, Spatial learning and memory Wei et al. (2018)
i.g., once a day for deficit, fear memory deficit
21 days apoptosis and loss of neuron ↑;
Bcl-2/Bax ratio ↑
122 In vitro PC-12 cells 5, 10, and 20 μmol/L Bcl-2 mRNA and protein Wei et al. (2018)
expressions ↑; Bax mRNA and
protein expressions ↓;
caspase-3 and caspase-9
expressions ↑; Bcl-2/Bax
ratio ↑
122 In vitro SH-SY5Y cells induced by 5, 10, and 20 μmol/L cell viability loss and apoptosis Lu et al. (2020)
Aβ1-42 ↓; Bax, caspase-3, caspase-8
and caspase-9 protein
expression levels ↓; Bcl-2
protein expression ↑
Reducing UVB-induced skin damage
EFAC In vitro Human HaCaT keratinocytes 50, 100, and 250 µg/ml Apoptotic cells number ↓; Ronpirin et al. (2016)
AFAC induced by UVB caspase 3 expression ↓; CPD ↓

NM, not mentioned; ACL, A. carambola leaves; MEACL, methanol extract of A. carambola leaves; EEBAC, ethanol extracts of bark from A. carambola; REAC, root extracts of A.
carambola; FPAC, fruit powder of A. carambola; JEAC, juice extracts of A. carambola; TFACL, total flavones from A. carambola leaf; EEACB, ethanol extracts of A. carambola bark;
MEACL, methanolic extract of A. carambola leaves; CEPAC, crude extract from peel of A. carambola; PEFAC, a homogenous polysaccharide extracted from the fruit of A. carambola; ACE,
A. carambola extracts; EEACL, ethanol extract of A. carambola leaves; FPEAC, free phenolic extract from A. carambola; FJAC, fruit juice of A. carambola; EACR, extract of A. carambola
root; AEAC, aqueous extract of A. carambola; AELAC, aqueous extract of leaves of A. carambola; EEACR, ethanol extracts of A. carambola root; FACF, flavonoids in A. carambola fruit;
EFAC, ethanol fractions of A. carambola; AFAC, aqueous fractions of A. carambola; 40, Norathyriol; 47, Carambolaflavone A; 48, Carambolaflavone B; 105, (+)-lyoniresinol 3α-O-β-D-
glucopyranoside; 106, (−)-lyoniresinol 3α-O-β-D-glucopyranoside; 121, 2-methoxy-6-nonyl-cyclohexa-2,5-diene-1,4-dione; 122, 2-dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione.

proteins, such as CTGF and TGF-β1, in kidney tissues markedly 100 mg/kg EEACB triggered a higher blood glucose level
decreased after JEAC treatment (Pham et al., 2017). JEAC (at decline than 50 mg/kg EEACB (Islam et al., 2020). These
daily doses of 5, 10, and 20 g/kg, i.g. for 14 days) has also been results suggest that A. carambola is a potential hypoglycemic
proven to considerably decrease FBG levels, blood glucose, area drug for the prevention and treatment of diabetes.
under the curve, LDH, glucagon (GC), and pyruvate in serum and
increase fasting insulin (FINS) levels in STZ-induced diabetic 6.2.2 Isolated Compounds
mice, indicating that this extract could lessen hyperglycemia and As previously mentioned, some compounds, including 47, 48,
hyperlipidemia, suppressing DN progression and development; it 100, 101, 120, and 121, isolated and identified from A. carambola
is a potential candidate agent for treating or preventing DN (Yang have been tested for their potential anti-hyperglycemic activity in
et al., 2019a). vivo. Normal hyperglycemic rats receiving an oral treatment of
Liu et al. (2013) reported that the total flavones from A. flavonoid carambolaflavone A (at doses of 20 and 50 mg/kg) (47)
carambola leaf (TFACL at daily doses of 0.2, 0.4, and 0.8 g/kg, and carambolaflavone B (at doses of 20 and 50 mg/kg) (48)
i.g. for 7 days) significantly lowered FBG levels and enhanced extracted from A. carambola leaves demonstrated decreased
glucose tolerance in mice and rats with diabetes mellitus. The glycemia levels, suggesting a potential hypoglycemic effect.
STZ-induced diabetic mice also received ethanol extracts of A. Glycogen levels in the muscle and liver also increased sharply
carambola bark (EEACB) orally (at daily doses of 50 and (Cazarolli et al., 2012). Another study by the same authors also
100 mg/kg for three successive days), which drastically found 50 mg/kg of carambolaflavone B (48) to notably decrease
decreased their level of blood glucose 150 min after orally blood glucose levels in diabetic rats and provoke glucose-
receiving a single dose of glucose solution (1.0 g/kg); triggered insulin secretion after oral treatment of the

Frontiers in Pharmacology | www.frontiersin.org 15 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

FIGURE 4 | Schematic representation of the underlying mechanism of anti-hyperglycemic activity of crude extracts or isolated compounds from A. carambola.

hyperglycemic rats. Additionally, a noteworthy stimulatory induced hyperglycemia, and the underlying molecular
function of compound 48 (at concentrations of 50 and mechanism of its impact could be associated with the
100 μM) on 14C-glucose uptake was observed, but treatment suppression of the activation of the TLR4/NF-κB signaling
with inhibitors, including wortmannin, RO318220, and pathway.
PD98059, reversed this activity. Interestingly, 100 μM of Zheng et al. (2013), Zheng et al. (2021) reported that DMDD
compound 48 and 10 nM of insulin had no synergistic effect (122) isolated from A. carambola dry roots (at daily doses of 12.5,
on glucose uptake (Cazarolli et al., 2009). 25, and 50 mg/kg, i.g. for 56 days) to significantly reduce FBG,
Chiral lignan glucosides, (±)-lyoniresinol 3α-O-β-D- creatinine clearance, proteinuria, Scr, and serum urea-N contents
glucopyranoside (LGP1 (105) and LGP2 (106)), isolated from and glomerular mesangial matrix expansion. It also markedly
A. carambola root have been evaluated for health-promoting improved renal AGE formation, decreased AGE receptor, NF-κB,
properties in LGP1 and LGP2-mediated hypoglycemia against TGF-β1, and Nε-(carboxymethyl)lysine expressions in diabetic
renal injury in STZ-created diabetic mice. Diabetic mice that mice, and effectively alleviated DN in type 2 diabetic KKAy mice.
received LGP1 and LGP2 (at daily doses of 20, 40, and 80 mg/kg However, the underlying mechanism of how DMDD alleviates
for 14 days) yielded contrasting outcomes. While LGP1 markedly DN has yet to be demonstrated; therefore, the precise action
alleviated the histopathological changes in the kidney and mechanism of DMDD against DN must be explored further (Lu
lowered FBG, FINS, and insulin sensitivity index (ISI) levels et al., 2019). STZ-established DN in TLR4 knockout (TLR4−/−,
and caspase-3/8/9, Bax, and NF-κB protein expressions, LGP2 KO) and wild-type (WT) mice treated orally with DMDD (at
had no impact (Wen et al., 2013), suggesting that LGP1 could doses of 12.5, 25, and 50 mg/kg for 28 days) lowered serum TC,
attenuate and treat the progression of DN through regulating TG, HDL-C, and LDL-C, blood glucose content and kidney
several molecular targets. function markers, including Scr and BUN, significantly and
Benzoquinone (121) isolated from the roots of A. carambola increased the quantity and density of podocytes spectacularly,
has also been analyzed, and, at doses of 30, 60, and 120 mg/kg, i.g. contributing to DN symptom alleviation. The treatment also
once a day for 21 days. It effectively decreased serum FBG, TC, markedly inhibited IL-6 and TNF-α levels and blocked the
TG, FFA, MCP-1, TNF-α, GHb, FINS, MDA, and IL-6 levels and TLR4/MyD88/NF-κB signaling pathways. These effects,
increased SOD and GSH activities in STZ-induced diabetic rats. though, were notably different in TLR4−/− mice. In vitro,
Mechanistically, the extract strikingly downregulated TNF-α, IL- 30 μM DMDD radically subdued TLR4, Smad2, and Smad3
6, MyD88, p-NF-κB, and TLR4 expressions in pancreatic tissues expressions in high glucose (HG)-stimulated HK-2 cells and
(Qin et al., 2019; Qin et al., 2020). These findings suggest that diminished BMP and activin membrane-bound inhibitor
benzoquinone (121) exerts a protective effect against STZ- (BAMBI) expressions (Zhang et al., 2019). Additionally, the

Frontiers in Pharmacology | www.frontiersin.org 16 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

FIGURE 5 | Schematic representation of the underlying mechanism of anti-tumor activity of crude extracts or isolated compounds from A. carambola.

increase in Smad2/3 expression and decrease in BAMBI reductions in urinary albumin, TC, TG, LDL-C, FBG, and CysC
expression in HG cultured cells were markedly annulled in levels. The extract also alleviated pathological changes and renal
cells treated with TAK-242 (a TLR4 signaling inhibitor). More fibrosis clearly but silencing the TLR4 gene improved the
importantly, BAMBI gene silencing extraordinarily enhanced the pathology. Mechanistically, TLR4, TGF-β1, Smad2, and
epithelial-mesenchymal transition (EMT) process and its over- Smad3 protein expressions in renal tissues decreased
expression did the opposite in HK-2 cells under HG conditions. pointedly after DMDD treatment. TGF-β1 and Smad2/3
EMT was eased with the pre-administration of DMDD to HK-2 genes and protein expressions also unusually declined in
cells, pointing to DMDD’s protective effects on HG-induced TLR4−/− mice than Wild type mice. Furthermore, according
EMT in HK-2 cells via suppressing the TLR4/BAMBI/Smad2/ to IHC results, there were strong in situ expressions of TLR4,
3 signaling pathways (Zhang et al., 2019). These findings TGF-β1, Smad2, and Smad3 in kidney tissues, which were
insinuate that DMDD isolated from A. carambola is a distinctly weakened after DMDD-treatment. Nevertheless,
potential therapeutic drug for DN treatment. TGF-β1, Smad2, and Smad3 levels did not increase in the
In Zhang et al. (2020) study of the antidiabetic activity of TLR4−/− mice with significance when compared with healthy
DMDD isolated from A. carambola roots in Wild type and TLR4 mice (Zhang et al., 2020). These findings strongly suggest that
knockout mice with diabetic kidney disease, DMDD (at daily TLR4 is critical for DMDD’s protective activity against renal
doses of 12.5, 25, and 50 mg/kg, i.g. for 28 days) caused notable insufficiency in diabetic mice, and hypoglycemic and anti-

Frontiers in Pharmacology | www.frontiersin.org 17 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

fibrosis properties could be regulated by the TLR4/TGF chronic diseases (Zhu et al., 2015; Li et al., 2016). One study found
signaling pathway. that some phytochemicals suppressed adipogenesis and obesity.
In summary, A. carambola could attenuate oxidative injury According to Rashid et al. (2016), the crude extract from the peel
and inflammatory response in rat models by enhancing of A. carambola (CEPAC) (at concentrations of 10, 100, 500, and
antioxidant enzyme activities and decreasing inflammatory 1,000 mg/ml) dose-dependently inhibited TG accumulation in
cytokine levels, which result in insulin secretion, as well as their research. Additionally, 1,000 mg/ml of CEPAC substantially
blood glucose and blood lipid metabolism regulation. Some blocked most of the adipocyte differentiation in 3T3-L1
original findings on active compounds and their related preadipocytes with no toxicity. The extract also notably
mechanisms also exist; however, these investigations lack clear reduced the mRNA expressions of two master adipogenic
indications of whether these activities stem from a single transcription factors, PPAR-γ and C/EBPα, and increased the
substance or synergistic effects of several substances of A. PPAR-α receptor at the molecular level. In further phytochemical
carambola; therefore, before they can be advanced as bases for analyses, the authors recognized (-)-epicatechin as a natural
clinical research and exploration of the medicinal values of A. bioactive molecule responsible for the features presented above
carambola, further evaluations must be performed. (Rashid et al., 2016). These findings indicate that CEPAC is
potentially promising in treating obesity and obesity-related
Anti-Hyperlipidemic Activity disorders.
Cardiovascular diseases (CVDs) are among the most common Per Li et al. (2016) inquiry into the benefits of DMDD against
causes of death globally, with hyperlipidemia representing one of high-fat diet (HFD)-evoked obesity and insulin resistance in
its main risk factors (Mahdavi et al., 2020; Jayachandran and Qu, mice, DMDD (at daily doses of 12.5, 25, and 50 mg/kg, i.g. for
2021). In Herman-Lara et al. (2014) evaluation of the protective 28 days) markedly decreased body and adipose tissue weights and
activity of micronized insoluble fiber from the bagasse of A. reduced insulin, blood glucose, TC, TG, FFA, IL-6, and TNF-α
carambola as an ingredient of a functional food (FF) or as levels in serum. DMDD also significantly downregulated TLR4
micronized insoluble fiber-rich fraction (IFRF) and its effects and MyD88 protein levels in epididymal adipose tissues,
in vivo on lipid metabolism in a murine model, serum TG, TC, considerably enhanced insulin secretion, lowered the areas
HDL-C, and LDL-C decreased after a murine model was treated under the curve, notably heightened SOD and GSH-Px
with IFRF and FF, with corresponding inhibition rates of 14.2, activities, and decreased MDA content in mice liver tissues (Li
25.4, 55.06, and 12.18% by IFRF, and 30.18, 39.47, 35.11, and et al., 2016). Based on these results, DMDD possesses potential
43.18% by FF. IFRF treatment also produced higher benefits that could be used to treat HFD-induced obesity and
hypolipidemic activity and greatly avoided the occurrence of insulin resistance, potentially improving the lipid metabolism
non-alcoholic fatty liver. Overall, IFRF and FF exerted process and the suppression of TLR4 protein expression in
hypolipidemic qualities, indicating that the star fruit’s adipose tissues (Table 2).
insoluble fiber is potentially a component of FFs in the
treatment and prevention of CVDs (Herman-Lara et al., 2014). Antitumor Activity
In Saghir et al. (2016) investigation of the anti-hyperlipidemic Several studies have demonstrated the significant inhibitory
activity of methanolic and aqueous extracts from various A. activity of A. carambola against a variety of tumor cells in vivo
carambola parts (such as leaves, stems, and ripe and unripe and in vitro and the possible mechanism of the antitumor activity
fruits), they found that the methanolic extract of A. carambola of crude extracts or isolated compounds from A. carambola
leaves (MEACL) had the most potent hypolipidemic effect in (Table 2 and Figure 5). DENA-induced and CCl4-promoted
poloxamer-407-induced hyperlipidemic rats and effectively liver cancer mice given A. carambola extracts (ACE, at a dose of
decreased serum TC, TG, LDL-C, VLDL-C, and atherogenic 25 mg/kg for 5 days) orally resulted in ACE considerably
index (AI) levels. MEACL (at daily doses of 250, 500, and lowering tumor incidence, tumor yield, and tumor burden.
1,000 mg/kg, i.g. for 35 days) significantly reduced serum and The treatment also significantly reduced lipid peroxidation
liver lipids levels, including TC, TG, LDL-C, VLDL-C, and AI, (LPO) levels and elevated GSH, SOD, CAT activities (Singh
elevated HDL-C, and dose-dependently alleviated et al., 2014). These findings hinted at ACE’s ability to exert a
histopathological changes in the liver of HFD-induced protective effect against liver cancer in mice and could be used as
hyperlipidemic rats. MEACL also considerably enhanced GSH, a good natural chemo-preventive product against cancer to
GSH-Px, SOD, and CAT activities and lowered MDA levels. further screen active components. Supposedly, MEACL (25
Furthermore, MEACL treatment inhibited HMG-CoA reductase and 50 mg/kg, i.g. once a day for 5 days) significantly reduces
and lipase (Aladaileh et al., 2019). These results demonstrate that body weight and cell viability, prolongs survival time, and
MEACL has a hypolipidemic activity it could exert by improves altered hematological parameters (HGB, WBC, RBC)
ameliorating lipid peroxidation and improving antioxidant in Ehrlich ascites carcinoma (EAC) cell-bearing mice. MELA
defenses in HFD-fed rats (Table 2). could induce cancer cell apoptosis through upregulating p53 and
Bax protein expressions at the molecular level (Siddika et al.,
Anti-Obesity Activity 2020).
Obesity-related diseases are increasingly becoming common Gao et al. (2015) also showed that DMDD significantly
public health and social issues. They can cause a multitude of suppressed breast cancer MCF-7 and BT20 cell growth, with
metabolic disorders closely associated with increased risk of many the IC50 values ranging from 3.13 to 5.57 μM; DMDD exerted

Frontiers in Pharmacology | www.frontiersin.org 18 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

anticancer traits by inducing apoptosis and blocking the cell cycle 0.03–1.0 mg/ear) shrunk the edema dose-dependently, with an
at the G1 phase, increasing the generation of intracellular ROS, ID50 value of 0.05 at concentrations of 0.02–0.13 mg/ear and
which activated the extrinsic receptor and intrinsic mitochondrial maximum inhibition of 73% at a concentration of 0.6 mg/ear.
pathways and inhibited NF-κB activation. One study using 4T1 EEACL also suppressed myeloperoxidase (MPO) activity, with an
breast cancer cell-bearing mice for in vivo research revealed that ID50 value of 0.22 and a maximum inhibition of 61% at a
DMDD radically inhibited primary breast tumor growth and concentration of 0.6 mg/ear. Furthermore, all the fractions
suppressed breast tumor metastasis in the lung and liver, as well examined markedly curbed edema formation and lowered
as decreased inflammatory cytokine production, induced cell MPO activity. Additional evaluations found that the ethyl
apoptosis, and prevented the activation of the TNF-α/NF-κB/ acetate fraction of EEACL was the most effective inhibitor of
MMPs pathways, prolonging the survival time of tumor-bearing MPO activity and edema formation, with inhibition rates of 54
mice (Chen et al., 2017b). These outcomes imply that DMDD has and 75%, respectively (Cabrini et al., 2011). Unfortunately,
potent antitumor features that could help treat metastatic breast EEACL bioactive substances against skin inflammation have
cancer. not been tested extensively so far; therefore, they require
Muhammed et al. (2019) explored the radio-sensitivity of 4T1 further scrutiny to be optimized and developed into effective
tumor cells treated with DMDD extracted from A. carambola remedies for preventing and treating skin disorders.
roots, by analyzing the TIE and TRD of the in vivo 2 and 24 h GC-MS analyses of a homogenous polysaccharide extracted
treatment groups compared with the untreated control group. from the fruit of A. carambola (PFSCW, Mw 40 kDa) established
The threshold ionization energy (TIE) and threshold radiation that it consisted mainly of arabinose, galactose, and galacturonic
dose (TRD) was determined utilizing a novel method that acid in a molar ratio of 12.3:1.7:86.0. Methylation and NMR
employs a laser trapping technique for single and multiple cell spectroscopy examination identified a substituted galacturonan
ionizations. Per the findings, treatment periods increased, and composed of (1→4)-linked-α-D-Galp A units with a branch at O-
TIE and TRD notably decreased, pointing to DMDD’s powerful 2 by (1→5)-linked-α-L-Araf and terminal-α-L-Araf and α-D-
enhancement effect on 4T1 cell radio-sensitivity. However, TRD Galp A units as components of PFSCW. PFSCW treatment (at
diminished with mass regardless of the intervention. TRD doses of 100 and 300 mg/kg) drastically reduced intraplantar
analyses of single vs multiple cells ionizations within each formalin-injected paw edema, with an inhibition rate of 53% at a
group continuously demonstrated the same behavior even dose of 300 mg/kg, suggesting that the extract has moderate anti-
after treatment. The underlying elements for these observed inflammatory properties (Leivas et al., 2016a). Xie et al. (2016)
relations could be explained in terms of radiation, demonstrated that DMDD (at concentrations of 10, 15, and
hyperthermia, and chemo effects (Muhammed et al., 2019). 20 μM) significantly inhibited palmitic acid (PA)-stimulated
DMDD treatment (at 4.0, 6.0, and 8.0 μg/ml for 24 h) has inflammation in pancreatic Min6 cells by strongly obstructing
been shown to dramatically and dose-dependently inhibit the the TLR4/MyD88/NF-KB pathways, which lowered the
proliferation, migration, and invasion of H1299 cells, with generation of inflammatory cytokines (TNF-α, IL-6, and MCP-
inhibition rates of 22.50, 30.13, and 58.87%, respectively. The 1), downregulating cleaved-caspase-3/8/9 protein expression,
same research determined that DMDD also induced the elevating the Bcl-2/Bax ratio, and decreasing TLR4, MyD88,
apoptosis of H1299 cells, with the mechanism possibly and NF-KB protein expressions. These outcomes hint at
associated with the suppression of the ERK/MAPK pathway DMDD’s ability to reverse PA-stimulated Min6 cell
(Zhou et al., 2019). dysfunction via alleviating cell apoptosis and inflammatory
Wu et al. (2020a) assessed the antitumor effect of DMDD on response, with the underlying mechanism of action almost
hepatocarcinoma in nude mice and its mechanism, the tumor certainly associated with the suppression of the TLR4/MyD88/
weight in mice decreased significantly, with tumor inhibition NF-κB signaling pathways (Table 2).
rates for the three doses amounting to 66.39, 63.11, and 47.33%,
respectively, after daily oral treatment with DMDD at 25, 50, and Hepatoprotective Activity
100 mg/kg for 12 days. The tumor and organ indexes, including An evaluation of the hepatic steatosis alleviating activity of a free
liver and spleen, and IL-2 and IL-10 serum levels also notably phenolic extract of A. carambola (FPEAC) in leptin receptor-
decreased, and the amounts of WBC, HGB, and PLT dramatically deficient (db/db) mice to clarify the underlying mechanisms of
increased. Additionally, the expression of TLR4, MyD88, and NF- action for modulating hepatic lipogenesis found that FPEAC (at
κB reduced markedly. These results suggest that DMDD has daily doses of 10, 20, and 30 g/kg, i.g. for 8 weeks) considerably
promising anticancer properties, with its mechanism of action is lowered LDL-C, TG, TC, non-esterified fatty acid (NEFA) levels,
possibly linked to the repression of the TLR4/MyD88/NF-κB and AST and ALT activities, elevated serum HDL-C level, and
signaling pathways (Table 2). reduced TG content in the liver. Mechanically, FPEAC drastically
downregulated sterol regulatory element-binding protein-1c
Anti-Inflammatory Activity (SREBP-1c), fatty acid synthase (FAS), and stearoyl-CoA
The evaluation of the anti-inflammatory effects of the ethanol desaturase 1 (SCD1) expressions in hepatic tissues and
extract of A. carambola leaves (EEACL), its ethyl acetate, butanol, markedly upregulated p-AMPK α levels. More importantly,
and hexane fractions, and two flavonoids against skin FPEAC significantly downregulated microRNA-34a and
inflammation in mice with croton oil-stimulated ear edema microRNA-33 expressions, which regulate the AMPK/SREBP-
revealed that topically used EEACL (at concentrations of 1c/FAS signaling pathway (Pang et al., 2017). These findings

Frontiers in Pharmacology | www.frontiersin.org 19 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

indicate that FPEAC has a compelling hepatic steatosis mitigating 25, and 50, 100 mg/kg/day, i.g., for 21 days) markedly alleviated
effect, partially through suppressing the signal transmission of the myocardial tissues damage, inhibited the myocardial cell
hepatic lipogenesis. According to Yang et al. (2018), the fruit juice apoptosis, reduced the levels of FBG, LVEDP, ROS, MDA,
of A. carambola (FJAC, at daily doses of 5, 10, and 20 g/kg, i.g. for Beclin-1, LC3II/I, and Atg5 as well as increased the SOD,
14 days) showed potential hepatoprotective properties; it reduced p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR, indicating that
MAD and cAMP levels and increased SDH, MDA, and SOD DMDD exerts cardioprotective activity via regulating the ROS-
activities in the liver of mice with STZ-induced diabetes. mediated PI3K/Akt/mTOR autophagy pathways (Table 2; Ma
In one study, mice with acute liver injury given the extract of et al., 2021).
A. carambola roots (EACR, at daily doses of 0.3, 0.6, and 1.2 g/kg
for 7 days) had significantly lower serum CCl4, AST, ALT, IL-1, Anti-Hypertensive Activity
and IL-6 levels and liver MDA levels but considerably increased In Soncini et al. (2011) study of the anti-hypertensive effect of the
SOD, GSH, and GSH-Px activities. At the molecular level, TNF-α, aqueous extract of leaves of A. carambola (AELAC) in an isolated
NF-κB, and caspase-3 protein expressions were significantly rat aorta and its possible mechanism, AELAC treatment (at doses
downregulated in the liver. HE staining showed that the liver of 12.5–50.0 mg/kg, i.v.) resulted in a remarkable dose-dependent
injury was eased (Huang et al., 2019). In another investigation, decrease in mean arterial pressure (MAP) in normotensive rats.
EACR (at daily doses of 0.25, 0.5, and 1.0 g/kg, i.g. for 28 days) In vitro, AELAC reduced Emax response to phenylephrine but
substantially increased the albumin/globulin (A/G) ratio, reduced triggered no sensitivity change. AEAC also suppressed CaCl2-
total bilirubin (TBIL) and TC levels in the liver microstructure of stimulated aorta contractions and led to the rightward shift of the
rats with CCl4-induced liver fibrosis (HF), markedly lessened NF- response curves of a depolarized Ca2+-free medium, suggesting
κB and Bax protein expressions, and considerably augmented that the extract repressed extracellular Ca2+ influx and caused
Bcl-2 protein expression (Liang et al., 2020b). Rats with CCl4- vasoconstriction. These outcomes strongly support the
instigated chronic liver injury treated intragastrically with EACR traditional use of A. carambola leaves in hypertension.
(at doses of 0.25, 0.5, and 1.0 g/kg for 56 days) displayed Orally administered ethanol extract of A. carambola roots
drastically decreased serum AST, ALT, AKP, and Hyp content, (EEACR, at 150, 300, and 600 mg/kg) has been shown to lower
serum hepatic fibrosis biomarkers (HA, LN, Col III, and Col IV), blood pressure in healthy rats at 300 and 600 mg/kg but have no
and liver tissue MDA content and increased SOD and GSH-Px noticeable effect on the heart rate (Tang et al., 2017). When
capacities. EACR also considerably reversed the elevation in Huang et al. (2017) explored the effect of flavonoids from A.
COL-1a1, α-SMA, TIMP2, TGF-β1, and Smad-2 and -4 carambola fruit (FACF) on healthy rats and rats with NG-nitro-
mRNA expressions and inhibited α-SMA, TIMP2, TGF-β1, L-arginine-methyl ester (L-NAME)-induced high blood pressure,
Smad-2, -3, -4, Bax, and caspase-3 levels in liver tissues. they established that FACF (at daily doses of 300, 600, and
Additionally, the extract significantly increased Smad-7 mRNA 1,200 mg/kg, i.g. for 5 weeks) significantly lessened systolic
expression and Smad-7 and Bcl-2 protein levels in the liver blood pressure, diastolic blood pressure, and mean blood
(Huang et al., 2020). These findings point to EACR being a pressure in healthy rats. Moreover, the blood pressure of rats
promising remedy for liver fibrosis. However, further studies with hypertension was also drastically lowered by 600 and
must be conducted urgently to identify the bioactive components 1,200 mg/kg FAC treatments, suggesting that FACF contains
and possible mechanism of EACR’s anti-fibrotic effects (Table 2). an active ingredient that decreases blood pressure (Table 2;
Huang et al., 2017).
Cardioprotective Activity
Ventricular remodeling (VR) results in changes in endothelial Neuroprotective Activity
vasoactive substances, cardiomyocyte hypertrophy, myocardial Alzheimer’s disease (AD), characterized by the progressive
fibrosis, and endothelial dysfunction. Per Liang et al. (2020a) deterioration of learning, memory, and cognition, is the most
investigation of the protective property of the aqueous extract of common, irreversible, and progressive neurodegenerative disease
A. carambola (AEAC) on isoprenaline-stimulated endothelial (Robins Wahlin and Byrne, 2011; Levenson et al., 2014). DMDD’s
function in rats with VR, AEAC (at daily doses of 50, 100, neuroprotective effect against memory deficits and neuron apoptosis
and 200 mg/kg, i.g. for 14 days) glaringly lowered serum levels in APP/PS1 transgenic AD mice has been reported (Wei et al., 2018).
of iNOS, TGF-β, Ang II, ECE, and ET-1 and their protein That research showed that mice receiving DMDD (at daily doses of
expressions and decreased the VR index and CVF but 12.5, 25, and 50 mg/kg for 21 days) orally displayed significantly
markedly elevated serum tNOS and eNOS levels and their improved memory and spatial learning and inhibited neuron loss
protein expressions. Pathological evaluations demonstrated and apoptosis in APP/PS1 hippocampal tissues. In vitro, DMDD (at
that AEAC radically alleviated inflammatory infiltration, concentrations of 5, 10, and 20 μmol/L) radically suppressed Aβ1-
apoptosis, fibrosis, and necrosis in rat myocardial tissues, 42-stimulated apoptosis by upregulating Bcl-2 expression and
suggesting that AEAC potentially mitigates the VR of rats and downregulating Bax expression, increasing the mitochondria
is, therefore, possibly associated with safeguarding the balance of membrane potential (MMP) and activating PC-12 cell caspase-3
vasoactive components and vascular endothelium function and caspase-9. Mice pretreated with DMDD also had drastically
(Liang et al., 2020a). Another study showed that the high- increased PC-12 cell Bcl-2/Bax ratio in vitro and in vivo (Wei et al.,
glucose-high-fat diet combined with STZ induced the diabetes 2018). These findings indicated that DMDD has beneficial
mellitus mice model administrated with DMDD (at doses of 12.5, properties against the deficit of learning and memory in APP/

Frontiers in Pharmacology | www.frontiersin.org 20 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

PS1 transgenic AD mice through improving neuron apoptosis and pyogenes, Staphylococcus saprophyticus, Streptococcus agalactiae, and
suppressing Bax/Bcl-2-mediated loss of MMP (Table 2). Pseudomonas spp. (Hossain et al., 2017).
Recently, Lu et al. (2020) explored the neuroprotective activity Recently, Silva et al. (2021) found that the extracts of stem
of DMDD against Aβ1-42-evoked SH-SY5Y cell apoptosis and the bark, leaves, and fruits from A. carambola perceptibly suppressed
underlying mechanism of DMDD’s protective function. Treatment the growth of multi-resistant pathogenic bacteria and fungi with a
with DMDD (at concentrations of 5, 10, and 20 μmol/L) minimal inhibitory concentration (MIC) of 100 μg/ml. Crude
significantly increased cell viability and inhibited Aβ1-42- extracts of the leaf exhibited a broad spectrum of action against
induced SH-SY5Y cell apoptosis. Mechanistically, DMDD Gram-positive and Gram-negative bacteria, such as S. aureus 10
markedly downregulated Bax, caspase-3, caspase-8, and caspase- MRSA, S. aureus ATCC 29213 MRSA, S. aureus 12 MRSA, S.
9 expressions, upregulated Bcl-2 levels, suppressed the release of aureus 6 MRSA, K. pneumoniae 8 ESBL, E. faecalis ATCC 29212,
cytochrome c, and the loss of MMP, and elevated the Bcl-2/Bax and A. baumannii 2 MBL (Silva et al., 2021). Overall, the findings
ratio (Lu et al., 2020). Overall, DMDD displayed excellent of these studies provide a research direction that points to A.
neuroprotective properties and is a promising ingredient for carambola’s prospective therapeutic efficacy against bacterial and
developing a neuroprotective drug for Alzheimer’s disease. fungal infections.

Reducing Ultraviolet B-Induced Skin TOXICITY ASSESSMENT


Damage
Ultraviolet B (UVB) is a major causation factor of cell injury and Literature material on the assessment of the toxicity and safety of
skin cancer. It causes DNA damage, promotes apoptosis, and A. carambola is limited, although this plant is commonly utilized
produces ROS. In Ronpirin et al. (2016) investigation of the in TCM to treat numerous ailments. In recent years, several
protective effect of A. carambola on HaCaT keratinocytes caused studies have found that the excessive consumption of A.
by UVB, the ethanol and aqueous fractions of A. carambola carambola results in toxic effects. Aranguren et al. (2017)
(EFAC and AFAC, at a concentration of 250 µg/ml) significantly literature review revealed that 123 patients from eight nations
decreased cell apoptosis. Both fractions also ominously lowered developed acute renal injury after consuming this fruit. Forty-
caspase 3 protein expression. Additionally, EFAC (at seven of the cases were registered in Brazil, the highest reported
concentrations of 100 and 250 µg/ml) and AFAC (at incidence, followed by Taiwan (36), Bangladesh (20), China and
concentrations of 50, 100, and 250 µg/ml) drastically reduced France (8 each), Sri Lanka (2), and Thailand and Colombia (1
the percentage of cyclobutane pyrimidine dimers (CPD), each); 28 of the patients died. In the most recent investigation,
resulting in DNA repair (Ronpirin et al., 2016). These reports Yasawardene et al. (2021) analyses of 10 case series and 28 case
suggest that A. carambola extracts could be developed reports in humans (total number of individuals  136) and eight
cosmetically to protect against UVB-induced skin damage animal studies for A. carambola nephrotoxicity and
(Table 2). neurotoxicity, 94 (69.1%) patients had prior renal impairment,
with renal histology revealing acute oxalate nephropathy with
Antimicrobial Activity tubulointerstitial nephritis or tubular necrosis. Also,
The extract of the stem bark, leaves, and fruits of A. carambola neurotoxicity performances ranged from hiccups to status
supposedly display antibacterial and antifungal activity. Previous epilepticus. The excessive uptake of A. carambola could lead
investigations demonstrated that the extracts of A. carambola fruits to acute kidney injury, particularly in an empty stomach, or
could suppress the growth of Staphylococcus aureus and Klebsiella dehydration state, and the use of A. carambola as therapy for an
spp., with the MBC of 15.62 and 125 mg/ml, respectively (Chang elderly patient is not recommended on an empty stomach
et al., 2000). Moreover, two compounds p-anisaldehyde and (Wijayaratne et al., 2018). In addition, A. carambola has
β-sitosterol (400 μg/disk) isolated from the bark of A. carambola threatened people’s health despite the relatively low frequency
strongly inhibited the growth of Escherichia coli with a zone of of star fruit intake. Further clinical studies on the toxic
inhibition of 15 mm and had moderate inhibitory activity against ingredients, metabolites, and intake dose of star fruit toxicity
fungi (Mia et al., 2007). Afterward, Wakte and Patil (2011) screened must be carried out to provide clear indications of intake and
the antimicrobial activity of fruit extracts of star fruit at various stages prevent toxicity and mortality.
against two Gram-positive bacteria (S. aureus ATCC 6538P and B. The results of animal experiments showed that the high
cereus ATCC 11778) and three Gram-negative bacteria (E. coli dose of the different A. carambola extracts showed different
ATCC 25922, P. aeruginosa ATCC 19429, and S. typhimurium degrees of toxicity or adverse reactions. Chen et al. (2001)
ATCC 23564), the results found that all the extracts show different found that an overdose intake of the fruit could result in
degrees of activity against Gram-positive and Gram-negative nephrotoxicity and neurotoxicity in some individuals (Chen
bacteria, and the methanol and acetone extracts were et al., 2001). Supposedly, the fruit of A. carambola could
considerably more effective than other solvent extracts in cause fatality in patients suffering from uremia, with oxalate
inhibiting the Gram-positive micro-organisms better than Gram identified as being a significant element in the toxicity of the
negative microorganisms. In addition, the ethanolic extracts of A. fruit (Dembitsky et al., 2011). Furthermore, an acute toxicity
carambola leaf at doses of 250 and 500 µg/disc were found to study conducted in female albino rats revealed that oral
moderately suppress the growth of Shigella dysenteriae, Streptococcus administration of A. carambola juice extract (ACJE) at

Frontiers in Pharmacology | www.frontiersin.org 21 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

different doses (250, 500, 1,000, 2000, 4,000, and well as various health and pharmacological actions of this fruit.
5,000 mg/kg) was safe and did not lead to any poisonous Phytochemical investigations have shown that 132 compounds
reactions after 48 h of treatment, despite the dosage reaching have been mainly reported from A. carambola. The flavonoids
up to 5,000 mg/kg. In the subacute evaluations, oral ACJE (at represented by compounds 47 and 48 and benzoquinones
dosages of 200, 400, and 600 mg/kg for 28 consecutive days) represented by DMDD have been considered as the
treatment elicited no significant difference in total protein, biologically active components with extensive biological
albumin, hematological parameters, and globulin values properties, including anti-hyperglycemic, anti-hyperlipidemic,
between treatment-receiving and control groups. However, anti-obesity, anti-inflammatory, hepatoprotective, anti-tumor,
serum AST, ALT, and ALP levels and urea, creatinine, and cardioprotective, and neuroprotective activities. Numerous
MDA levels in the treat-receiving group dose-dependently studies have revealed that A. carambola might be a promising
increased significantly more than in the control group. candidate to treat diabetes mellitus and DN-related diseases.
Moreover, when compared with the control group, the Furthermore, A. carambola can be employed in the prevention
histomorphology of the livers and kidneys of the rats and treatment of ailments related to aging and oxidative stress. In
treated with ACJE displayed lesions of degeneration and summary, A. carambola, as a food and medicinal resource, has a
necrosis (Aba and Amadi, 2019). Recent investigations good health care function and important edible and medicinal
systematically revealed that the nephrotoxic effect of A. value, and thus has good prospects for utilization.
carambola stems primarily from oxalate deposition in There are nevertheless still many problems in research that
renal tubules, causing interstitial nephritis and acute aims to bridge the gap between health and the bioactive
tubular necrosis (Yasawardene et al., 2020). These results substances of A. carambola, which need to be studied
indicate that excessive and long-term consumption of ACJE further: firstly, although there are a large number of in vitro
could be nephrotoxic and hepatotoxic. Therefore, it is very and in vivo studies concentrated on the crude extracts of A.
important to study the intake dose as well as effective and safe carambola using exorbitant doses, to the best of our
dose in the future. knowledge, the active substances from these crude extracts
Phytochemical studies have shown that A. carambola contains with bioactivities are still unknown. Thus, research aiming to
two poisonous substances, caramboxin, and oxalic acid. clarify the biological activities and the mechanism of active
Caramboxin is a non-proteinogenic amino acid that stimulates components found in A. carambola should be clearly
the glutamate receptors in neurons. The chemical structure of conducted in further depth, as they will certainly
caramboxin is similar to the amino acid phenylalanine, it is collaborate to the establishment of safe dosages and
metabolized and excreted through the kidneys (Yasawardene accelerate the steps for the discovery of new molecules of
et al., 2020). Caramboxin can effectively stimulate the central biological interest. Secondly, the bioactive compounds and
nervous system (CNS), resulting in symptoms of CNS disorder, crude extracts of A. carambola have been increasingly and
including mental confusion, seizures, and status epilepticus successfully utilized in diabetic disease prevention and health
(Yasawardene et al., 2020). It can cause belch, vomiting, promotion in the last few decades. The documentary evidence
confusion, consciousness disorders, and shock, etc. If normal points to a high number of bioactive compounds and crude
people eat this fruit, caramboxin can be safely discharged, so extracts in star fruit that might be used as functional food
normal people will not be damaged by the toxin when it is sources against various illnesses including diabetes, cancer,
ingested. However, the patients with renal insufficiency, cardiovascular disease, and other oxidative stresses or aging-
especially in patients undergoing peritoneal dialysis or induced chronic diseases. There is a need for further research
hemodialysis can’t eat it. Huynh and Nguyen (2017) findings to explore and utilize the processed products of this fruit,
showed that the alcohol fermentation technologies efficaciously especially those phenolic extracts and dietary fibers in A.
reduced oxalate contents in A. carambola, preventing the risk of carambola for functional food formulation. Thirdly,
kidney stone formation. However, the works on the dose of although toxicity studies of A. carambola have been
caramboxin and the bioavailability of oxalate and caramboxin reported, the information related to its toxicity mechanism,
after ingestion are still insufficient, and more comprehensive, especially nephrotoxicity and neurotoxicity, is still lacking. It is
systematic, and authentic studies are required to assess the dose of reported that A. carambola induced oxalate nephropathy
caramboxin of star fruit juice and its pharmacokinetic parameters remains an under-recognized cause for both acute and
in healthy individuals. chronic kidney disease. Therefore, more public awareness
about oxalate poisoning on uremic patients should be
promoted. This will help to avoid adverse reactions to star-
FUTURE OUTLOOKS fruits in high uremic patients. The public must be well
educated on the benefits as well as the hazardous effects of
This review summarized the botany, ethnopharmacology, star fruits. Furthermore, toxicological studies are crucial for
phytochemistry, pharmacology, and toxicity of A. carambola understanding the safety of herbal drugs. Therefore, to ensure
to explore this valuable fruit. The succulence and sweet taste the full use of its medicinal resources, further acute toxicity,
of starfruit are of interest to the food industry. The and subacute toxicity, as well as the safety assessment studies
pharmaceutical and health industries have been increasingly of A. carambola, both in vitro and in vivo, should be carried
interested in A. carambola due to the nutritional properties as out. Fourthly, continued efforts will be needed to clarify the

Frontiers in Pharmacology | www.frontiersin.org 22 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

pathways of pharmacokinetics including absorption, AUTHOR CONTRIBUTIONS


distribution, metabolism, and excretion, and to assess the
long-term chronic toxicity and acute toxicity as well as the FL obtained the literatures, wrote the manuscript, and finalized
metabolites of phytochemicals formed in vivo of the bioactive the paper. LP and ZL arranged the tables and designed the figures,
compounds, especially for DMDD before proceeding to the XJ collected the pictures. JZ, YY, and XH retrieved relevant
development of pharmaceutical formulation. The literature and discussed the manuscript layout. NZ and XH
pharmacokinetics parameters also contribute to facilitate the designed and revised the manuscript, checked the accuracy,
rational optimization of natural active substances, and and edited the final version. All authors read and approved
increase therapeutic effects and reduce toxicities. Overall, the final version of the manuscript for publication.
in-depth research on clinical studies to justify their reported
therapeutic potential, clinical efficacy, and safety of A.
carambola is imminent. FUNDING
Taken together, significant interest has been generated over A.
carambola owing to the numerous beneficial effects. A. carambola This study was financially supported by the National Natural
appears to have great developing potential in the fields of Science Foundation of China (Grant No. 82074094), the Open
functional food and modern medicine. Therefore, the Research Fund of Chengdu University of Traditional Chinese
opportunities and challenges co-exist. Meanwhile, it is an Medicine Key Laboratory of Systematic Research of Distinctive
effective approach for TCM to get into the international Chinese Medicine Resources in Southwest China (Grant No.
market and an ideal choice for modern health care in 2020XSGG002), the Xinglin Scholar Research Promotion
developed countries for ongoing human health and thereby Project of Chengdu University of Traditional Chinese Medicine
building a healthy community with a shared future for (Grant No. CDTD2018014), the Science and Technology Plan
mankind. We believe that A. carambola will have an extensive Project of Guizhou Province (Grant No. Qian KeHe Platform
international market and a broad prospect in terms of its Talents (2018)5772-074), and the Science and Technology Project
applications in both medicine and functional food. of Zunyi (Grant No. ZSKH-HZ-(2020)-78).

Carolino, R., Beleboni, R., Pizzo, A., Vecchio, F., Garciacairasco, N., Moysesneto,
REFERENCES M., et al. (2005). Convulsant Activity and Neurochemical Alterations Induced
by a Fraction Obtained from Fruit (Oxalidaceae: Geraniales). Neurochem. Int.
Aba, P. E., and Amadi, A. U. (2020). Evaluation of the Possible Hepatotoxic and 46 (7), 523–531. doi:10.1016/j.neuint.2005.02.002
Nephrotoxic Potentials of the Averrhoa carambola Juice Extract in Female Albino Cazarolli, L. H., Folador, P., Moresco, H. H., Brighente, I. M. C., Pizzolatti, M. G.,
Rats. J. Basic Clin. Physiol. Pharmacol. 31 (1), 20190042. doi:10.1515/jbcpp-2019- and Silva, F. R. M. B. (2009). Mechanism of Action of the Stimulatory Effect of
0042 Apigenin-6-C-(2″-O-α-l-Rhamnopyranosyl)-β-L-Fucopyranoside on 14C-
Abeysekera, R. A., Wijetunge, S., Nanayakkara, N., Wazil, A. W., Ratnatunga, N. Glucose Uptake. Chem. Biol. Interact. 179, 407–412. doi:10.1016/
V., Jayalath, T., et al. (2015). Star Fruit Toxicity: a Cause of Both Acute Kidney j.cbi.2008.11.012
Injury and Chronic Kidney Disease: a Report of Two Cases. BMC Res. Notes 8, Cazarolli, L. H., Kappel, V. D., Pereira, D. F., Moresco, H. H., Brighente, I. M. C.,
796. doi:10.1186/s13104-015-1640-8 Pizzolatti, M. G., et al. (2012). Anti-hyperglycemic Action of Apigenin-6-C-
Aladaileh, S., Saghir, S., Murugesu, K., Sadikun, A., Ahmad, A., Kaur, G., et al. β-Fucopyranoside from Averrhoa Carambola. Fitoterapia 83, 1176–1183.
(2019). Antihyperlipidemic and Antioxidant Effects of Averrhoa Carambola doi:10.1016/j.fitote.2012.07.003
Extract in High-Fat Diet-Fed Rats. Biomedicines 7 (3), 72. doi:10.3390/ Chakthong, S., Chiraphan, C., Jundee, C., Chaowalit, P., and Voravuthikunchai, S.
biomedicines7030072 P. (2010). Alkyl Phenols from the wood of Averrhoa Carambola. Chin. Chem.
Araho, D., Miyakoshi, M., Chou, W. H., Kambara, T., Mizutani, K., and Ikeda, T. Lett. 21, 1094–1096. doi:10.1016/j.cclet.2010.03.031
(2005). A New Flavone C-Glycoside from the Leaves of Averrhoa Carambola. Chang, J. M., Hwang, S. J., Kuo, H. T., Tsai, J. C., Guh, J. Y., Chen, H. C., et al.
Nat. Med. 59, 113–116. doi:10.1016/j.apsusc.2004.03.089 (2000). Fatal Outcome after Ingestion of Star Fruit (Averrhoa carambola) in
Aranguren, C., Vergara, C., and Rosselli, D. (2017). Toxicity of Star Fruit (Averrhoa Uremic Patients. Am. J. Kidney Dis. 35 (2), 189–193. doi:10.1016/s0272-
carambola) in Renal Patients: A Systematic Review of the Literature. Saudi 6386(00)70325-8
J. Kidney Dis. Transpl. 28 (4), 709–715. Chen, C. L., Fang, H. C., Chou, K. J., Wang, J. S., and Chung, H. M. (2001). Acute
Bahramsoltani, R., Kalkhorani, M., Abbas Zaidi, S. M., Farzaei, M. H., and Rahimi, Oxalate Nephropathy after Ingestion of star Fruit. Am. J. Kidney Dis. 37 (2),
R. (2020). The Genus Tamarix: Traditional Uses, Phytochemistry, and 418–422. doi:10.1053/ajkd.2001.21333
Pharmacology. J. Ethnopharmacol. 246, 112245. doi:10.1016/j.jep.2019.112245 Chen, C. X., Nong, Z. H., Xie, Q. Q., He, J. H., Cai, W. E., Tang, X. N., et al. (2017b).
Benkeblia, N., and Lopez, M. G. (2015). Saccharides and Fructooligosaccharides 2-dodecyl-6-Methoxycyclohexa-2,5-Diene-1,4-Dione Inhibits the Growth and
Composition of Green and Ripe Averrhoa carambola, Blighia sapida and Spondias Metastasis of Breast Carcinoma in Mice. Sci. Rep. 7 (1), 6704. doi:10.1038/
dulcis Fruits. Food Chem. 176, 314–318. doi:10.1016/j.foodchem.2014.12.080 s41598-017-07162-3
Bhat, R., Ameran, S. B., Voon, H. C., Karim, A. A., and Tze, L. M. (2011). Quality Chen, G. L., Zhang, X., Chen, S. G., Han, M. D., and Gao, Y. Q. (2017a).
Attributes of Starfruit (Averrhoa carambola L.) Juice Treated with Ultraviolet Antioxidant Activities and Contents of Free, Esterified and Insoluble-Bound
Radiation. Food Chem. 127 (2), 641–644. doi:10.1016/j.foodchem.2011.01.042 Phenolics in 14 Subtropical Fruit Leaves Collected from the South of China.
Bircher, A. G., and Bircher, W. H. (2000). Encyclopedia of Fruit Trees and Edible J. Funct. Foods 30, 290–302. doi:10.1016/j.jff.2017.01.011
Flowering Plants in Egypt and the Subtropics. Cairo, Egypt: American University Chen, L., Lu, X., El-Seedi, H., and Teng, H. (2019). Recent Advances in
in Cairo Press, 596. the Development of Sesquiterpenoids in the Treatment of Type 2
Cabrini, D. A., Moresco, H. H., Imazu, P., da Silva, C. D., Pietrovski, E. F., Mendes, Diabetes. Trends Food Sci. Technol. 88, 46–56. doi:10.1016/
D. A., et al. (2011). Analysis of the Potential Topical Anti-inflammatory j.tifs.2019.02.003
Activity of Averrhoa carambola L. in Mice. Evid. Based Complement. Chua, C. B., Sun, C. K., Tsui, H. W., Yang, P. J., Lee, K. H., Hsu, C. W., et al. (2017).
Alternat Med. 2011, 908059. doi:10.1093/ecam/neq026 Association of Renal Function and Symptoms with Mortality in star Fruit

Frontiers in Pharmacology | www.frontiersin.org 23 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

(Averrhoa carambola) Intoxication. Clin. Toxicol. 55 (7), 624–628. doi:10.1080/ Lakmal, K., Yasawardene, P., Jayarajah, U., and Seneviratne, S. L. (2021).
15563650.2017.1314490 Nutritional and Medicinal Properties of Star Fruit (Averrhoa carambola L.):
Dembitsky, V. M., Poovarodom, S., Leontowicz, H., Leontowicz, M., Vearasilp, S., A Review. Food Sci. Nutr. 9 (3), 1810–1823. doi:10.1002/fsn3.2135
Trakhtenberg, S., et al. (2011). The Multiple Nutrition Properties of Some Leivas, C. L., Iacomini, M., and Cordeiro, L. M. C. (2016b). Pectic Type II
Exotic Fruits: Biological Activity and Active Metabolites. Food Res. Int. 44, Arabinogalactans from Starfruit (Averrhoa carambola L.). Food Chem. 199,
1671–1701. doi:10.1016/j.foodres.2011.03.003 252–257. doi:10.1016/j.foodchem.2015.12.020
Fan, Y. Y., Sahu, S. K., Yang, T., Mu, W. X., Wei, J. P., Cheng, L., et al. (2020). Leivas, C. L., Iacomini, M., and Cordeiro, L. M. C. (2015). Structural
Dissecting the Genome of star Fruit (Averrhoa carambola L.). Hortic. Res. 7, 94. Characterization of a Rhamnogalacturonan I-Arabinan-Type I
doi:10.1038/s41438-020-0306-4 Arabinogalactan Macromolecule from Starfruit (Averrhoa carambola L).
Farias, D. P., Neri-Numa, I. A., de Araújo, F. F., and Pastore, G. M. (2020). A Carbohydr. Polym. 121, 224–230. doi:10.1016/j.carbpol.2014.12.034
Critical Review of Some Fruit Trees from the Myrtaceae Family as Promising Leivas, C. L., Nascimento, L. F., Barros, W. M., Santos, A. R. S., Iacomini, M., and
Sources for Food Applications with Functional Claims. Food Chem. 306, Cordeiro, L. M. C. (2016a). Substituted Galacturonan from Starfruit: Chemical
125630. doi:10.1016/j.foodchem.2019.125630 Structure and Antinociceptive and Anti-Inflammatory Effects. Int. J. Biol.
Ferreira, E. B., Fernandes, L. C., Galende, S. B., Cortez, A. G., and Bazotte, R. B. Macromol. 84, 295–300. doi:10.1016/j.ijbiomac.2015.12.034
(2008). Hypoglycemic Effect of the Hydroalcoholic Extract of Leaves of Levenson, R. W., Sturm, V. E., and Haase, C. M. (2014). Emotional and Behavioral
Averrhoa carambola L. (Oxalidaceae). Rev. Bras. Farmacogn. 18 (3), Symptoms in Neurodegenerative Disease: a Model for Studying the Neural
339–343. doi:10.1590/s0102-695x2008000300005 Bases of Psychopathology. Annu. Rev. Clin. Psychol. 10, 581–606. doi:10.1146/
Flora of China Editorial Committee (1998). Flora of China. Beijing, China: Science annurev-clinpsy-032813-153653
Press. Li, J., Wei, X., Xie, Q., Hoa Pham, T. T., Wei, J., He, P., et al. (2016). Protective
Gao, Y., Huang, R., Gong, Y., Park, H. S., Wen, Q., Almosnid, N. M., et al. (2015). Effects of 2-Dodecyl-6-Methoxycyclohexa-2,5 -Diene-1,4-Dione Isolated from
The Antidiabetic Compound 2-Dodecyl-6-Methoxycyclohexa-2,5-Diene-1,4- Averrhoa Carambola L. (Oxalidaceae) Roots on High-Fat Diet-Induced Obesity
Dione, Isolated from Averrhoa carambola L., Demonstrates Significant and Insulin Resistance in Mice. Cell. Physiol. Biochem. 40 (5), 993–1004.
Antitumor Potential against Human Breast Cancer Cells. Oncotarget 6 (27), doi:10.1159/000453156
24304–24319. doi:10.18632/oncotarget.4475 Liang, C. X., Pham, H. T. T., Zhang, X. L., Liang, X. M., Huang, R. B., and Huang, X.
Gunasegaran, R. (1992). Flavonoids and Anthocyanins of Three Oxalidaceae. (2020b). Effect of Extract of Averrhoa carambola L. Root on Liver
Fitoterapia 63, 89–90 . Microstructure of Liver Fibrosis Rats. J. Guangxi Med. Univ. 37 (7),
Gunawardena, D. C., Jayasinghe, L., and Fujimoto, Y. (2015). Phytotoxic 1245–1249. doi:10.16190/j.cnki.45-1211/r.2020.07.008
Constituents of the Fruits of Averrhoa Carambola. Chem. Nat. Compd. 51 Liang, X., Huang, R., Huang, J., Chen, C., Qin, F., Liu, A., et al. (2020a). Effect of an
(3), 532–533. doi:10.1007/s10600-015-1332-6 Aqueous Extract of Averrhoa carambola L. on Endothelial Function in Rats
Herman-Lara, E., Elvira-Torales, L. I., Rodriguez-Miranda, J., Torruco-Uco, J. G., with Ventricular Remodelling. Biomed. Pharmacother. 121, 109612.
Carmona-García, R., Mendoza-García, P. G., et al. (2014). Impact of doi:10.1016/j.biopha.2019.109612
Micronized Starfruit (Averrhoa carambola L.) Fiber Concentrate on Lipid Liao, P. Y., Li, D. P., Hu, Z. Y., Chen, H. P., Yang, X. M., and Wu, Y. Y. (2019b).
Metabolism in Mice. Int. J. Food Sci. Nutr. 65 (7), 862–867. doi:10.3109/ Extraction Process Optimization and In Vitro Activity Evaluation for
09637486.2014.918590 Polysaccharides from Averrhoa carambola Roots. Chin. Tradit. Pat. Med. 41
Hossain, T., Barman, A. K., Karmakar, U. K., Bokshi, B., Dev, S., and Biswas, N. N. (9), 2030–2034. doi:10.3969/j.issn.1001-1528.2019.09.002
(2017). Phytochemical and Pharmacological Evaluation of Leaves of Averrhoa Liao, P. Y., Zhou, Z. Y., Chen, Y. R., Chen, H. P., Pan, W. G., Yang, X. M., et al.
carambola Linn. (Family: Oxalidaceae). Biosci. Bioeng. Commun. 3 (1), 144–151. (2019a). Chemical Constituents of the Water Extracts of Averrhoa carambola L.
Huang, W. S., Pham, H. T. T., Xuan, F. F., Li, J. M., and Huang, R. B. (2017). Effects Root and Their Biological Activities. Nat. Prod. Res. Dev. 31, 81–86.
of Flavonoids Compound in Averrhoa carambola L. (Oxalidacaeae) Fruit doi:10.16333/j.1001-6880.2019.1.013
(ACFTF) on Blood Pressure of Rats. China J. Tradit. Chin. Med. Pharm. 32 Liu, F. Z., Song, X. M., Wang, X. L., Liu, T., and Liang, R. F. (2013). Hypoglycemic
(4), 1786–1788. Effect of Total Flavones from Averrhoa carambola L. Leaf. China J. Tradit. Chin.
Huang, X., Wang, L., Meng, M., Zhang, S., Pham, T. T. H., Jiang, L., et al. (2020). Med. Pharm. 19 (11), 279–281.doi:10.11653/syfj2013110279
Extract of Averrhoa carambola L. (Oxalidaceae) Roots Ameliorates Carbon Lu, S. Y., Wei, X. J., Zhang, H. L., Chen, Z. F., Li, J. M., Xu, X. H., et al. (2020).
Tetrachloride-Induced Hepatic Fibrosis in Rats. Biomed. Pharmacother. 121, Protective Effect of 2-Dodecyl-6-Methoxycyclohexa-2, 5-Diene-1, 4-Dione,
109516. doi:10.1016/j.biopha.2019.109516 Isolated from Averrhoa carambola L., Against Aβ1-42-Induced Apoptosis in
Huang, X., Xie, Q. Q., Ye, F. X., Qin, L. H., Huang, R. B., and Zhang, S. J. (2019). SH-SY5Y Cells by Reversing Bcl-2/Bax Ratio. Psychopharmacology 238 (1),
Protective Effect of Extract of Averrhoa carambola L. Root on CCl4-Induced 193–200. doi:10.1007/s00213-020-05668-9
Acute Liver Injury in Mice. Chin. Pharmacol. Bull. 35 (1), 106–110. doi:10.3969/ Lu, S., Zhang, H., Wei, X., Huang, X., Chen, L., Jiang, L., et al. (2019). 2-Dodecyl-6-
j.issn.1001-1978.2019.01.021 Methoxycyclohexa-2,5-Diene-1,4-Dione Isolated from Averrhoa carambola L.
Huynh, N. K., and Nguyen, H. V. H. (2017). Effects of Juice Processing on Oxalate Root Ameliorates Diabetic Nephropathy by Inhibiting the TLR4/MyD88/NF-Κb
Contents in Carambola Juice Products. Plant Foods Hum. Nutr. 72 (3), Pathway. Diabetes Metab. Syndr. Obes. 12, 1355–1363. doi:10.2147/dmso.s209436
236–242. doi:10.1007/s11130-017-0615-4 Lu, Y., Tan, C. W., Chen, D., and Liu, S.-Q. (2018). Potential of Three Probiotic
Islam, S., Alam, M. B., Ahmed, A., Lee, S., Lee, S.-H., and Kim, S. (2020). Lactobacilli in Transforming Star Fruit Juice into Functional Beverages. Food
Identification of Secondary Metabolites in Averrhoa carambola L. Bark by Sci. Nutr. 6 (8), 2141–2150. doi:10.1002/fsn3.775
High-Resolution Mass Spectrometry and Evaluation for α-glucosidase, Ma, J., Wu, Y. N., Huang, D. L., Zhou, Y. P., Hou, L. T., Li, Y. K., et al. (2021).
Tyrosinase, Elastase, and Antioxidant Potential. Food Chem. 332, 127377. Averrhoa carambola L. Roots DMDD Alleviates Myocardial Injury in Diabetes
doi:10.1016/j.foodchem.2020.127377 Mellitus Mice by Regulating ROS-Mediated Autophagy Pathways. Chin.
Jayachandran, M., and Qu, S. (2021). Harnessing Hyperuricemia to Atherosclerosis Pharmacol. Bull. 37 (6), 823–827. doi:10.3969/j.issn.1001-1978.2021.06.015
and Understanding its Mechanistic Dependence. Med. Res. Rev. 41, 616–629. Mahdavi, A., Bagherniya, M., Fakheran, O., Reiner, Ž., Xu, S., and Sahebkar, A.
doi:10.1002/med.21742 (2020). Medicinal Plants and Bioactive Natural Compounds as Inhibitors of
Jia, X., Xie, H., Jiang, Y., and Wei, X. (2018). Flavonoids Isolated from the Fresh HMG-CoA Reductase: A Literature Review. Biofactors 46, 906–926.
Sweet Fruit of Averrhoa Carambola , Commonly Known as star Fruit. doi:10.1002/biof.1684
Phytochemistry 153, 156–162. doi:10.1016/j.phytochem.2018.06.007 Maqsood, S., Adiamo, O., Ahmad, M., and Mudgil, P. (2020). Bioactive Compounds
Jia, X., Yang, D., Xie, H., Jiang, Y., and Wei, X. (2017). Non-Flavonoid Phenolics from from Date Fruit and Seed as Potential Nutraceutical and Functional Food
Averrhoa carambola Fresh Fruit. J. Funct. Foods 32, 419–425. doi:10.1016/j.jff.2017.03.025 Ingredients. Food Chem. 308, 125522. doi:10.1016/j.foodchem.2019.125522
Jia, X., Yang, D., Yang, Y., and Xie, H. (2019). Carotenoid-Derived Flavor Mia, M., Rahman, M., Begum, K., Begum, B., and Rashid, M. (2007).
Precursors from Averrhoa carambola Fresh Fruit. Molecules 24, 256. Phytochemical and Biological Studies of Averrhoa carambola. Dhaka Univ.
doi:10.3390/molecules24020256 J. Pharm. Sci. 6, 125–128. doi:10.3329/dujps.v6i2.688

Frontiers in Pharmacology | www.frontiersin.org 24 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

Mohd Suhaimi, N. I., Mat Ropi, A. A., and Shaharuddin, S. (2021). Safety and Shui, G., and Leong, L. P. (2006). Residue from star Fruit as Valuable Source for
Quality Preservation of Starfruit (Averrhoa carambola) at Ambient Shelf Life Functional Food Ingredients and Antioxidant Nutraceuticals. Food Chem. 97,
Using Synergistic Pectin-Maltodextrin-Sodium Chloride Edible Coating. 277–284. doi:10.1016/j.foodchem.2005.03.048
Heliyon 7 (2), e06279. doi:10.1016/j.heliyon.2021.e06279 Siddika, A., Zahan, T., Khatun, L., Habib, M. R., Aziz, M. A., Tareq, A. R. M., et al.
Moresco, H. H., Queiroz, G. S., Pizzolatti, M. G., and Brighente, I. M. C. (2012). (2020). In Vivo the Antioxidative Extract of Averrhoa carambola Linn. Leaves
Chemical Constituents and Evaluation of the Toxic and Antioxidant Activities Induced Apoptosis in Ehrilch Ascites Carcinoma by Modulating P53 Expression.
of Averrhoa carambola Leaves. Rev. Bras. Farmacogn. 22, 319–324. doi:10.1590/ Food Sci. Biotechnol. 29 (9), 1251–1260. doi:10.1007/s10068-020-00775-x
s0102-695x2011005000217 Silva, K. B., Pinheiro, C. T. S., Soares, C. R. M., Souza, M. A., Matos-Rocha, T. J., Fonseca, S.
Muhammed, E., Chen, L., Gao, Y., and Erenso, D. (2019). Chemo-Treated 4T1 A., et al. (2021). Phytochemical Characterization, Antioxidant Potential and
Breast Cancer Cells Radiation Response Measured by Single and Multiple Cell Antimicrobial Activity of Averrhoa carambola L. (Oxalidaceae) against
Ionization Using Infrared Laser Trap. Sci. Rep. 9 (1), 17547. doi:10.1038/ Multiresistant Pathogens. Braz. J. Biol. 81 (3), 509–515. doi:10.1590/1519-6984.220259
s41598-019-53821-y Silva, K. D. R. R., and Sirasa, M. S. F. (2018). Antioxidant Properties of Selected
Muthu, N., Lee, S. Y., Lee, S. Y., Phua, K. K., and Bhore, S. J. (2016). Nutritional, Fruit Cultivars Grown in Sri Lanka. Food Chem. 238, 203–208. doi:10.1016/
Medicinal and Toxicological Attributes of Star-Fruits (Averrhoa carambola L.): j.foodchem.2016.08.102
A Review. Bioinformation 12 (12), 420–424. doi:10.6026/97320630012420 Singh, R., Sharma, J., and Goyal, P. K. (2014). Prophylactic Role of Averrhoa carambola
Pang, D., You, L., Li, T., Zhou, L., Sun-Waterhouse, D., and Liu, R. H. (2016). Phenolic (Star Fruit) Extract against Chemically Induced Hepatocellular Carcinoma in Swiss
Profiles and Chemical- or Cell-Based Antioxidant Activities of Four Star Fruit Albino Mice. Adv. Pharmacol. Sci. 2014, 158936. doi:10.1155/2014/158936
(Averrhoa carambola) Cultivars. RSC Adv. 6, 90646–90653. doi:10.1039/c6ra15692d Soncini, R., Santiago, M. B., Orlandi, L., Moraes, G. O. I., Peloso, A. L. M., dos
Pang, D., You, L., Zhou, L., Li, T., Zheng, B., and Liu, R. H. (2017). Averrhoa Carambolafree Santos, M. H., et al. (2011). Hypotensive Effect of Aqueous Extract of Averrhoa
Phenolic Extract Ameliorates Nonalcoholic Hepatic Steatosis by Modulating carambola L. (Oxalidaceae) in Rats: an In Vivo and In Vitro Approach.
mircoRNA-34a, mircoRNA-33 and AMPK Pathways in Leptin Receptor-Deficient J. Ethnopharmacol. 133 (2), 353–357. doi:10.1016/j.jep.2010.10.001
db/db Mice. Food Funct. 8 (12), 4496–4507. doi:10.1039/c7fo00833c Sritharan, T., Savitri Kumar, N., Jayasinghe, L., Araya, H., and Fujimoto, Y. (2019).
Pham, H. T., Huang, W., Han, C., Li, J., Xie, Q., Wei, J., et al. (2017). Effects of Averrhoa Isocoumarins and Dihydroisocoumarins from the Endophytic Fungus
carambola L. (Oxalidaceae) Juice Mediated on Hyperglycemia, Hyperlipidemia, and Biscogniauxia capnodes Isolated from the Fruits of Averrhoa carambola. Nat.
its Influence on Regulatory Protein Expression in the Injured Kidneys of Product. Commun. 14, 1934578X1985196. doi:10.1177/1934578X19851969
Streptozotocin-Induced Diabetic Mice. Am. J. Transl. Res. 9 (1), 36–49. Tang, J. Z., Nong, H. L., Liang, X. M., Huang, J. C., Qin, F. Z., and Huang, R. B.
Provesi, J. G., Valentim Neto, P. A., Arisi, A. C. M., and Amante, E. R. (2019). (2017). Effect of the Ethanol Extracts of Averrhoa carambola Root on Blood
Extraction of Antifreeze Proteins from Cold Acclimated Leaves of Drimys Pressure in Normal Rats. West. China J. Pharm. Sci. 32 (2), 160–162.
Angustifolia and Their Application to star Fruit (Averrhoa carambola) doi:10.1016/j.biopha.2019.109612
Freezing. Food Chem. 289, 65–73. doi:10.1016/j.foodchem.2019.03.055 Teng, H., Yuan, B., Gothai, S., Arulselvan, P., Song, X., and Chen, L. (2018). Dietary
Qin, L. H., Wu, X. C., Zhou, X., Wu, Y. N., Huang, R. B., and Zhang, S. J. (2019). Triterpenes in the Treatment of Type 2 Diabetes: To Date. Trends Food Sci.
Effects of Benzoquinone of Averrhoa carambola L. Root on Glucose and Lipid Technol. 72, 34–44. doi:10.1016/j.tifs.2017.11.012
Metabolism, Oxidative Stress and Inflammatory Injury in Diabetic Mice. Chin. Thomas, R., Jebin, N., Saha, R., and Sarma, D. K. (2016). Antioxidant and
Pharmacol. Bull. 35 (12), 1720–1725. doi:10.3969/j.issn.1001-1978.2019.12.019 Antimicrobial Effects of Kordoi (Averrhoa carambola) Fruit Juice and
Qin, L., Zhang, X., Zhou, X., Wu, X., Huang, X., Chen, M., et al. (2020). Protective Bamboo (Bambusa polymorpha) Shoot Extract in Pork Nuggets. Food
Effect of Benzoquinone Isolated from the Roots of Averrhoa carambola L. on Chem. 190, 41–49. doi:10.1016/j.foodchem.2015.05.070
Streptozotocin-Induced Diabetic Mice by Inhibiting the TLR4/NF-κB Signaling Uuh-Narváez, J. J., González-Tamayo, M. A., and Segura-Campos, M. R. (2021). A
Pathway. Diabetes Metab. Syndr. Obes. 13, 2129–2138. doi:10.2147/dmso.s241998 Study on Nutritional and Functional Study Properties of Mayan Plant Foods as
Ramadan, N. S., Wessjohann, L. A., Mocan, A., C Vodnar, D., H. El-Sayed, N., A. a New Proposal for Type 2 Diabetes Prevention. Food Chem. 341, 128247.
El-Toumy, S., et al. (2020). Nutrient and Sensory Metabolites Profiling of doi:10.1016/j.foodchem.2020.128247
Averrhoa carambola L. (Starfruit) in the Context of its Origin and Ripening Valim, F. d. P., Aguiar-Oliveira, E., Kamimura, E. S., Alves, V. D., and Maldonado,
Stage by GC/MS and Chemometric Analysis. Molecules 25 (10), 2423. R. R. (2016). Production of Star Fruit Alcoholic Fermented Beverage. Indian
doi:10.3390/molecules25102423 J. Microbiol. 56 (4), 476–481. doi:10.1007/s12088-016-0601-9
Rashid, A. M., Lu, K., Yip, Y. M., and Zhang, D. (2016). Averrhoa carambola L. Peel Varela-Martínez, D. A., González-Curbelo, M. Á., González-Sálamo, J., and
Extract Suppresses Adipocyte Differentiation in 3T3-L1 Cells. Food Funct. 7 (2), Hernández-Borges, J. (2019). High-Throughput Analysis of Pesticides in
881–892. doi:10.1039/c5fo01208b Minor Tropical Fruits from Colombia. Food Chem. 280, 221–230.
Robins Wahlin, T.-B., and Byrne, G. J. (2011). Personality Changes in Alzheimer’s doi:10.1016/j.foodchem.2018.12.045
Disease: a Systematic Review. Int. J. Geriat. Psychiatry 26 (10), 1019–1029. Vasant, R. A., and Narasimhacharya, A. V. R. L. (2014). Antidotal Activity of
doi:10.1002/gps.2655 Averrhoa carambola (Star Fruit) on Fluoride Induced Toxicity in Rats.
Ronpirin, C., Pattarachotanant, N., and Tencomnao, T. (2016). Protective Interdiscip. Toxicol. 7 (2), 103–110. doi:10.2478/intox-2014-0014
Effect of Mangifera indica Linn., Cocos nucifera Linn., and Averrhoa Vasconcelos, C. M. L., Araújo, M. S., and Conde-Garcia, E. A. (2006).
carambola Linn. Extracts against Ultraviolet B-Induced Damage in Electrophysiological Effects of the Aqueous Extract of Averrhoa carambola
Human Keratinocytes. Evid. Based Compl. Alternat. Med. 2016, L. Leaves on the Guinea Pig Heart. Phytomedicine 13 (7), 501–508. doi:10.1016/
1684794. doi:10.1155/2016/1684794 j.phymed.2005.01.013
Rufino, M. S. M., Alves, R. E., Fernandes, F. A. N., and Brito, E. S. (2011). Free Wakte, S. R., and Patil, D. A. (2011). Antimicrobial and Antioxidant Activity of
Radical Scavenging Behavior of Ten Exotic Tropical Fruits Extracts. Food Res. Averrhoa Carambola L. Fruit at Various Stages of Ripening. J. Herb. Med.
Int. 44 (7), 2072–2075. doi:10.1016/j.foodres.2010.07.002 Toxicol. 5 (2), 121–129.
Saghir, S. A., Sadikun, A., Al-Suede, F. S., Majid, A. M., and Murugaiyah, V. (2016). Wang, Y., Liu, M., Liu, L., Xia, J.-H., Du, Y.-G., and Sun, J.-S. (2018). The Structural
Antihyperlipidemic, Antioxidant and Cytotoxic Activities of Methanolic and Revision and Total Synthesis of Carambolaflavone A. J. Org. Chem. 83 (7),
Aqueous Extracts of Different Parts of Star Fruit. Curr. Pharm. Biotechnol. 17 4111–4118. doi:10.1021/acs.joc.8b00008
(10), 915–925. doi:10.2174/1389201017666160603013434 Wei, S.-D., Chen, H., Yan, T., Lin, Y.-M., and Zhou, H.-C. (2014). Identification of
Saikia, S., Mahnot, N. K., and Mahanta, C. L. (2015). Optimisation of Phenolic Antioxidant Components and Fatty Acid Profiles of the Leaves and Fruits from
Extraction from Averrhoa carambola Pomace by Response Surface Averrhoa carambola. LWT - Food Sci. Technol. 55, 278–285. doi:10.1016/j.lwt.2013.08.013
Methodology and its Microencapsulation by spray and Freeze Drying. Food Wei, X. J., Xu, X. H., Chen, Z. F., Liang, T., Wen, Q. W., Qin, N., et al. (2018).
Chem. 171, 144–152. doi:10.1016/j.foodchem.2014.08.064 Protective Effects of 2-Dodecyl-6-Methoxycyclohexa-2,5-Diene-1,4-Dione
Shui, G., and Leong, L. P. (2004). Analysis of Polyphenolic Antioxidants in star Isolated from Averrhoa carambola L. (Oxalidaceae) Roots on Neuron
Fruit Using Liquid Chromatography and Mass Spectrometry. J. Chromatogr. A Apoptosis and Memory Deficits in Alzheimer’s Disease. Cell. Physiol.
1022, 67–75. doi:10.1016/j.chroma.2003.09.055 Biochem. 49 (3), 1064–1073. doi:10.1159/000493289

Frontiers in Pharmacology | www.frontiersin.org 25 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

Wen, Q., Lin, X., Liu, Y., Xu, X., Liang, T., Zheng, N., et al. (2012). Phenolic and Fruit Juice. Food Sci. Dev. 39 (9), 148–151. doi:10.3969/j.issn.1005-
Lignan Glycosides from the Butanol Extract of Averrhoa carambola L. Root. 6521.2018.09.028
Molecules 17, 12330–12340. doi:10.3390/molecules171012330 Yang, Y. X., Huang, T. M., Huang, R. B., and Xu, X. H. (2019a). The Effects and
Wen, Q. W., Chen, C. X., Liang, X. M., Xu, X. H., and Huang, R. B. (2014). Isolation, Mechanism of Averrhoa carambola Fruit Juice on Blood Glucose in Diabetic Mice.
Identification and Determination for Benzoquinone Compounds from Averrhoa J. Guangxi Med. Univ. 36 (1), 7–10. doi:10.16190/j.cnki.45-1211/r.2019.01.002
carambola Root. Chin. J. Exp. Tradit. Med. Formulae 20 (11), 70–73. Yang, Y., Xie, H., Jiang, Y., and Wei, X. (2020b). Flavan-3-ols and 2-
doi:10.13422/j.cnki.syfjx.2014110070 Diglycosyloxybenzoates from the Leaves of Averrhoa carambola. Fitoterapia
Wen, Q. W., Liang, T., Qin, F. Z., Wei, J. B., He, Q. L., Luo, X., et al. (2013). Lyoniresinol 3α- 140, 104442. doi:10.1016/j.fitote.2019.104442
O-β-D-Glucopyranoside-Mediated Hypoglycaemia and its Influence on Apoptosis- Yao, Q., Gao, Y., Lai, C., Wu, C., Zhao, C. L., Wu, J. L., et al. (2020). The
Regulatory Protein Expression in the Injured Kidneys of Streptozotocin-Induced Mice. Phytochemistry, Pharmacology and Applications of Melicope pteleifolia: A
PLoS One 8 (12), e81772. doi:10.1371/journal.pone.0081772 Review. J. Ethnopharmacol. 251, 112546. doi:10.1016/j.jep.2020.112546
Wijayaratne, D. R., Bavanthan, V., de Silva, M. V. C., Nazar, A. L. M., and Yasawardene, P., Jayarajah, U., De Zoysa, I., and Seneviratne, S. L. (2020).
Wijewickrama, E. S. (2018). Star Fruit Nephrotoxicity: a Case Series and Mechanisms of Star Fruit (Averrhoa carambola) Toxicity: A Mini-Review.
Literature Review. BMC Nephrol. 19 (1), 288. doi:10.1186/s12882-018-1084-1 Toxicon 187, 198–202. doi:10.1016/j.toxicon.2020.09.010
World Health Organization and Regional Office for the Western Pacific (2007). Yasawardene, P., Jayarajah, U., De Zoysa, I., and Seneviratne, S. L. (2021).
WHO International Standard Terminologies on Traditional Medicine in the Nephrotoxicity and Neurotoxicity Following Star Fruit (Averrhoa
Western Pacific Region. Manila: WHO Regional Office for the Western Pacific. carambola) Ingestion: A Narrative Review. Trans. R. Soc. Trop. Med. Hyg.
Wu, S. S., Sun, W., Xu, Z. C., Zhai, J. W., Li, X. P., Li, C. R., et al. (2020b). The doi:10.1093/trstmh/trab026
Genome Sequence of Star Fruit (Averrhoa carambola). Hortic. Res. 7, 95. Zhang, H., Lu, S., Chen, L., Huang, X., Jiang, L., Li, Y., et al. (2020). 2-Dodecyl-6-
doi:10.1038/s41438-020-0307-3 Methoxycyclohexa-2,5-Diene-1,4-Dione, Isolated from the Root of Averrhoa
Wu, X. C., Lu, S. Y., Zhou, X., Qin, L. H., Jiang, L. H., Li, Y. C., et al. (2020a). Anti- carambola L., Protects against Diabetic Kidney Disease by Inhibiting TLR4/
hepatocarcinoma Effect of and its Mechanism of 2-Dodecyl-6- TGFβ Signaling Pathway. Int. Immunopharmacol. 80, 106120. doi:10.1016/
Methoxycyclohexa-2,5-Diene-1,4-Dione in Averrhoa carambola L. Roots. j.intimp.2019.106120
Chin. J. Hosp. Pharm. 40 (1), 42–47. doi:10.13286/j.1001-5213.2020.01.05 Zhang, H., Wei, X., Lu, S., Lin, X., Huang, J., Chen, L., et al. (2019). Protective Effect of
Xie, Q., Zhang, S., Chen, C., Li, J., Wei, X., Xu, X., et al. (2016). Protective Effect of DMDD, Isolated from the Root of Averrhoa carambola L., on High Glucose
2-Dodecyl-6-Methoxycyclohexa-2, 5-Diene-1, 4-Dione, Isolated from Induced EMT in HK-2 Cells by Inhibiting the TLR4-BAMBI-Smad2/3 Signaling
Averrhoa carambola L., Against Palmitic Acid-Induced Inflammation and Pathway. Biomed. Pharmacother. 113, 108705. doi:10.1016/j.biopha.2019.108705
Apoptosis in Min6 Cells by Inhibiting the TLR4-MyD88-NF-κB Signaling Zheng, N., Lin, X., Wen, Q., Kintoko, S., Zhang, J., et al. (2013). Effect of 2-Dodecyl-
Pathway. Cell. Physiol. Biochem. 39 (5), 1705–1715. doi:10.1159/000447871 6-Methoxycyclohexa-2,5-Diene-1,4-Dione, Isolated from Averrhoa carambola
Xu, X. H., Phama, H. T. T., Wei, X. J., Qin, N., and Huang, R. B. (2017). Study of the L. (Oxalidaceae) Roots, on Advanced Glycation End-Product-Mediated Renal
Extract of Averrhoa carambola L. Root on Renal Function in Diabetic Mice and Injury in Type 2 Diabetic KKAy Mice. Toxicol. Lett. 219 (1), 77–84. doi:10.1016/
its Anti-Oxidative Action. Chin. Pharmacol. Bull. 33 (1), 95–100. doi:10.3969/ j.toxlet.2013.03.001
j.issn.1001-1978.2017.01.017 Zheng, N., Lin, X., Wen, Q., Kintoko, S., Zhang, J., et al. (2021). Effect of 2-Dodecyl-
Xu, X., Liang, T., Lin, X., Wen, Q., Liang, X., Li, W., et al. (2015). Effect of the Total 6-Methoxycyclohexa-2, 5-Diene-1, 4-Dione, Isolated from Averrhoa carambola
Extract of Averrhoacarambola (Oxalidaceae) Root on the Expression Levels of L. (Oxalidaceae) Roots, on Advanced Glycation End-Product-Mediated Renal
TLR4 and NF-κB in Streptozotocin-Induced Diabetic Mice. Cell. Physiol. Injury in Type 2 Diabetic KKAy Mice. Toxicol. Lett. 339, 88–96. doi:10.1016/
Biochem. 36 (6), 2307–2316. doi:10.1159/000430194 j.toxlet.2020.11.022
Xu, X., Liang, T., Wen, Q., Lin, X., Tang, J., Zuo, Q., et al. (2014). Protective Effects Zhou, X., Qin, L. H., Wu, X. C., Wu, Y. N., Chen, L. X., and Huang, R. B. (2019).
of Total Extracts of Averrhoa carambola L. (Oxalidaceae) Roots on Inhibition Effect of 2-Dodecyl-6-Methoxycyclohexa-2,5-Diene-1,4-Dione on
Streptozotocin-Induced Diabetic Mice. Cell. Physiol. Biochem. 33 (5), Lung Cancer H1299 Cells. Chin. J. Clin. Pharmacol. 35 (21), 2675–2678.
1272–1282. doi:10.1159/000358695 Zhu, C., Xiao, Y., Liu, X., Han, J., Zhang, J., Wei, L., et al. (2015). Pioglitazone
Yang, D.-P., Liu, X., Teng, C. P., Owh, C., Win, K. Y., Lin, M., et al. (2017). Lowers Serum Retinol Binding Protein 4 by Suppressing its Expression in
Unexpected Formation of Gold Nanoflowers by a Green Synthesis Method as Adipose Tissue of Obese Rats. Cell. Physiol. Biochem. 35 (2), 778–788.
Agents for a Safe and Effective Photothermal Therapy. Nanoscale 9 (41), doi:10.1159/000369737
15753–15759. doi:10.1039/c7nr06286a Zulfajri, M., Dayalan, S., Li, W. Y., Chang, C. J., Chang, Y. P., and Huang, G. G.
Yang, D., Jia, X., and Xie, H. (2019b). Heptyl Vicianoside and Methyl Caramboside (2019). Nitrogen-Doped Carbon Dots from Averrhoa carambola Fruit Extract
from Sour star Fruit. Nat. Product. Res. 33 (8), 1233–1236. doi:10.1080/ as a Fluorescent Probe for Methyl Orange. Sensors 19 (22), 5008. doi:10.3390/
14786419.2018.1466123 s19225008
Yang, D., Jia, X., Xie, H., and Wei, X. (2016). Further Dihydrochalcone
C-Glycosides from the Fruit of Averrhoa carambola. LWT - Food Sci. Conflict of Interest: The authors declare that the research was conducted in the
Technol. 65, 604–609. doi:10.1016/j.lwt.2015.08.061 absence of any commercial or financial relationships that could be construed as a
Yang, D., Xie, H., Jia, X., and Wei, X. (2015). Flavonoid C-Glycosides from Star potential conflict of interest.
Fruit and Their Antioxidant Activity. J. Funct. Foods 16, 204–210. doi:10.1016/
j.jff.2015.04.048 Publisher’s Note: All claims expressed in this article are solely those of the authors
Yang, D., Xie, H., Yang, B., and Wei, X. (2014). Two Tetrahydroisoquinoline and do not necessarily represent those of their affiliated organizations, or those of
Alkaloids from the Fruit of Averrhoa carambola. Phytochem. Lett. 7, 217–220. the publisher, the editors and the reviewers. Any product that may be evaluated in
doi:10.1016/j.phytol.2013.12.007 this article, or claim that may be made by its manufacturer, is not guaranteed or
Yang, L., Zhu, J., Song, L., Shi, X., Li, X., and Yu, R. (2012). Three Sesquiterpene endorsed by the publisher.
Compounds Biosynthesised from Artemisinic Acid Using Suspension-Cultured
Cells of Averrhoa carambola (Oxalidaceae). Nat. Product. Res. 26 (15), Copyright © 2021 Luan, Peng, Lei, Jia, Zou, Yang, He and Zeng. This is an open-
1388–1394. doi:10.1080/14786419.2011.589055 access article distributed under the terms of the Creative Commons Attribution
Yang, Y., Jia, X., Xie, H., and Wei, X. (2020a). Dihydrochalcone C-Glycosides from License (CC BY). The use, distribution or reproduction in other forums is permitted,
Averrhoa carambola Leaves. Phytochemistry 174, 112364. doi:10.1016/ provided the original author(s) and the copyright owner(s) are credited and that the
j.phytochem.2020.112364 original publication in this journal is cited, in accordance with accepted academic
Yang, Y. X., Huang, T. M., Huang, R. B., and Xu, X. H. (2018). Study on the Liver practice. No use, distribution or reproduction is permitted which does not comply
Tissue Anti-Oxidative Stress and Hypoglycemic Effects of Averrhoa carambola with these terms.

Frontiers in Pharmacology | www.frontiersin.org 26 August 2021 | Volume 12 | Article 699899


Luan et al. Phytochemistry and Pharmacology of Averrhoa carambola

GLOSSARY GSH reduced glutathione


GSH-Px glutathione peroxidase
ACP acid phosphatase HA hyaluronic acid
AI atherogenic index HDL-C high-density lipoprotein cholesterol
AIP atherogenic index of plasma Hyp hydroxyproline
AKP alkaline phosphatase iNOS inducible NO synthase
ALP alkaline phosphatase IL-6 interleukin-6
ALT alanine aminotransferase ISI insulin sensitivity index
AMPK α AMP-activated protein kinase α LDL-C low-density lipoprotein cholesterol
Ang II angiotensin II LN laminin
AST aspartate aminotransferase LPO lipid peroxidation
BAMBI BMP and activin membrane bound inhibitor MDA malondialdehyde
Bax bcl-associated X protein MPO myeloperoxidase
BUN blood urea nitrogen Myd88 myeloid differentiation factor 88
cAMP cyclic adenosine monophosphate NEFA non-esterified fatty acid
CAT catalase NF-κB nuclear factor-κB
C/EBPα CCAAT-enhancer binding protein α p-AMPK α phospho-AMPK α
Col III collagen type III PPAR-α/γ peroxisome proliferator activated receptors-α/γ
Col IV collagen type IV Scr serum creatinine
CTGF connective tissue growth factor SCD1 stearoyl-CoA desaturase 1
CVDs cardiovascular diseases SDH sorbitol dehydrogenase
CVF collagen volume fraction SGOT serum glutamate oxaloacetate transaminase
CysC cystatin C SGPT serum pyruvate transaminase
DENA diethylnitrosamine SOD superoxide dismutase
DMDD 2-dodecyl-6-methoxycyclohexa-2,5-diene- 1,4-dione SREBP-1c sterol regulatory element binding protein-1c
DPPH 2,2-diphenyl-1-picrylhydrazyl radical scavenging activity STZ streptozotocin
eNOS endothelial NO synthase TAA total ascorbic acid
ECE endothelin-converting enzyme TC total cholesterol
ET-1 endothelin 1 TEAC trolox equivalent antioxidant capacity
FAS fatty acid synthase TG triglycerides
FBG fasting blood glucose TGF-β1 transforming growth factor beta 1
FFA free fatty acids TL total lipids
FINS fasting insulin TLR4 Toll-like receptors-4
FRAP ferric reducing antioxidant power tNOS total NO synthase
G-6-Pase glucose-6-phosphatase TNF-α tumor necrosis factor-α
GC-MS gas chromatography mass spectrometry TQ triglyceride
GHb glucosylated hemoglobin VLDL-C very low-density lipoprotein cholesterol

Frontiers in Pharmacology | www.frontiersin.org 27 August 2021 | Volume 12 | Article 699899

You might also like